1
|
Grahovac J, Đurić A, Tanić M, Krivokuća A. Sex-Related Differences in Pancreatic Ductal Adenocarcinoma Progression and Response to Therapy. Int J Mol Sci 2024; 25:12669. [PMID: 39684385 DOI: 10.3390/ijms252312669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly malignancies with an increasing incidence rate and limited therapeutic options. Biological sex has an impact on many aspects of PDAC development and response to therapy, yet it is highly unappreciated in both basic and translational research, and worryingly in PDAC clinical trials. In this review, we summarize how biological sex influences PDAC incidence and mortality, genetic and epigenetic landscapes, anti-tumor immunity, responses to hormones, cachexia, and the efficacy of therapy. We highlight the importance of sex as a variable and discuss how to implement it into preclinical and clinical research. These considerations should be of use to researchers aiming at improving understanding of PDAC biology and developing precision medicine therapeutic strategies.
Collapse
Affiliation(s)
- Jelena Grahovac
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Đurić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Miljana Tanić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Krivokuća
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
2
|
Branch C, Parson-Martinez J, Cory TJ. Drug-drug interactions in HIV-infected patients receiving chemotherapy. Expert Opin Drug Metab Toxicol 2024:1-13. [PMID: 39305240 DOI: 10.1080/17425255.2024.2408004] [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: 04/30/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Coadministration of antiretrovirals and anti-cancer medications may present many complex clinical scenarios. This is characterized by the potential for drug-drug interactions (DDIs) and the challenges that arise in patient management. In this article, we investigate the potential for DDIs between antiretrovirals, including protease inhibitors (PIs), non-nucleoside reverse transcriptase inhibitors, nucleoside reverse transcriptase inhibitors (NRTIs), integrase strand transfer inhibitors (INSTIs), and anti-cancer medications. AREAS COVERED PubMed, Google Scholar, and Clinicaltrials.gov were searched for relevant articles in April 2024. Our review highlights PIs and NNRTIs as particularly prone to DDIs with anticancer agents, with implications for efficacy and toxicity of concomitant cancer therapy. We explain the mechanisms for interactions, emphasizing the significance of pharmacokinetic effects and enzyme induction or inhibition. We discuss clinical challenges encountered in the management of patients receiving combined ART and cancer therapy regimens. EXPERT OPINION Data are lacking for potential DDIs between antiretroviral and anti-cancer agents. While some interactions are documented, others are theoretical and based on the pharmacokinetic properties of the medications. Awareness of these interactions, inter-collaborative care between healthcare providers, and standardized treatment guidelines are all crucial for achieving optimal treatment outcomes and ensuring the well-being of patients with HIV/AIDS and cancer comorbidities.
Collapse
Affiliation(s)
- Chrystalyn Branch
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, TN, USA
| | - Jan Parson-Martinez
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, TN, USA
| | - Theodore James Cory
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, TN, USA
| |
Collapse
|
3
|
Lucchetti M, Aina KO, Grandmougin L, Jäger C, Pérez Escriva P, Letellier E, Mosig AS, Wilmes P. An Organ-on-Chip Platform for Simulating Drug Metabolism Along the Gut-Liver Axis. Adv Healthc Mater 2024; 13:e2303943. [PMID: 38452399 DOI: 10.1002/adhm.202303943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/26/2024] [Indexed: 03/09/2024]
Abstract
The human microbiome significantly influences drug metabolism through the gut-liver axis, leading to modified drug responses and potential toxicity. Due to the complex nature of the human gut environment, the understanding of microbiome-driven impacts on these processes is limited. To address this, a multiorgan-on-a-chip (MOoC) platform that combines the human microbial-crosstalk (HuMiX) gut-on-chip (GoC) and the Dynamic42 liver-on-chip (LoC), mimicking the bidirectional interconnection between the gut and liver known as the gut-liver axis, is introduced. This platform supports the viability and functionality of intestinal and liver cells. In a proof-of-concept study, the metabolism of irinotecan, a widely used colorectal cancer drug, is imitated within the MOoC. Utilizing liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS), irinotecan metabolites are tracked, confirming the platform's ability to represent drug metabolism along the gut-liver axis. Further, using the authors' gut-liver platform, it is shown that the colorectal cancer-associated gut bacterium, Escherichia coli, modifies irinotecan metabolism through the transformation of its inactive metabolite SN-38G into its toxic metabolite SN-38. This platform serves as a robust tool for investigating the intricate interplay between gut microbes and pharmaceuticals, offering a representative alternative to animal models and providing novel drug development strategies.
Collapse
Affiliation(s)
- Mara Lucchetti
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
| | | | - Léa Grandmougin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
| | - Christian Jäger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
| | - Pau Pérez Escriva
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belval, L-4362, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belval, L-4362, Luxembourg
| | - Alexander S Mosig
- Institute of Biochemistry II, Jena University Hospital, D-07747, Jena, Germany
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belval, L-4362, Luxembourg
| |
Collapse
|
4
|
Picher EA, Wahajuddin M, Barth S, Chisholm J, Shipley J, Pors K. The Capacity of Drug-Metabolising Enzymes in Modulating the Therapeutic Efficacy of Drugs to Treat Rhabdomyosarcoma. Cancers (Basel) 2024; 16:1012. [PMID: 38473371 DOI: 10.3390/cancers16051012] [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: 01/16/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Rhabdomyosarcoma (RMS) is a rare soft tissue sarcoma (STS) that predominantly affects children and teenagers. It is the most common STS in children (40%) and accounts for 5-8% of total childhood malignancies. Apart from surgery and radiotherapy in eligible patients, standard chemotherapy is the only therapeutic option clinically available for RMS patients. While survival rates for this childhood cancer have considerably improved over the last few decades for low-risk and intermediate-risk cases, the mortality rate remains exceptionally high in high-risk RMS patients with recurrent and/or metastatic disease. The intensification of chemotherapeutic protocols in advanced-stage RMS has historically induced aggravated toxicity with only very modest therapeutic gain. In this review, we critically analyse what has been achieved so far in RMS therapy and provide insight into how a diverse group of drug-metabolising enzymes (DMEs) possess the capacity to modify the clinical efficacy of chemotherapy. We provide suggestions for new therapeutic strategies that exploit the presence of DMEs for prodrug activation, targeted chemotherapy that does not rely on DMEs, and RMS-molecular-subtype-targeted therapies that have the potential to enter clinical evaluation.
Collapse
Affiliation(s)
- Enric Arasanz Picher
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Julia Chisholm
- Children and Young People's Unit, Royal Marsden Hospital, Institute of Cancer Research, Sutton SM2 5PR, UK
| | - Janet Shipley
- Sarcoma Molecular Pathology Group, Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Klaus Pors
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
5
|
Joyce SA, Clarke DJ. Microbial metabolites as modulators of host physiology. Adv Microb Physiol 2024; 84:83-133. [PMID: 38821635 DOI: 10.1016/bs.ampbs.2023.12.001] [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: 06/02/2024]
Abstract
The gut microbiota is increasingly recognised as a key player in influencing human health and changes in the gut microbiota have been strongly linked with many non-communicable conditions in humans such as type 2 diabetes, obesity and cardiovascular disease. However, characterising the molecular mechanisms that underpin these associations remains an important challenge for researchers. The gut microbiota is a complex microbial community that acts as a metabolic interface to transform ingested food (and other xenobiotics) into metabolites that are detected in the host faeces, urine and blood. Many of these metabolites are only produced by microbes and there is accumulating evidence to suggest that these microbe-specific metabolites do act as effectors to influence human physiology. For example, the gut microbiota can digest dietary complex polysaccharides (such as fibre) into short-chain fatty acids (SCFA) such as acetate, propionate and butyrate that have a pervasive role in host physiology from nutrition to immune function. In this review we will outline our current understanding of the role of some key microbial metabolites, such as SCFA, indole and bile acids, in human health. Whilst many studies linking microbial metabolites with human health are correlative we will try to highlight examples where genetic evidence is available to support a specific role for a microbial metabolite in host health and well-being.
Collapse
Affiliation(s)
- Susan A Joyce
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - David J Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland.
| |
Collapse
|
6
|
Žunec S, Karačonji IB, Čatalinac M, Jurič A, Katić A, Kozina G, Micek V, Neuberg M, Vrdoljak AL. Effects of concomitant use of THC and irinotecan on tumour growth and biochemical markers in a syngeneic mouse model of colon cancer. Arh Hig Rada Toksikol 2023; 74:198-206. [PMID: 37791673 PMCID: PMC10549892 DOI: 10.2478/aiht-2023-74-3765] [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: 07/01/2023] [Revised: 07/01/2023] [Accepted: 09/01/2023] [Indexed: 10/05/2023] Open
Abstract
Clinical treatment with the antineoplastic drug irinotecan (IRI) is often hindered by side effects that significantly reduce the quality of life of treated patients. Due to the growing public support for products with Δ9-tetrahydrocannabinol (THC), even though relevant scientific literature does not provide clear evidence of their high antitumour potential, some cancer patients take unregistered preparations containing up to 80 % THC. This study was conducted on a syngeneic colorectal cancer mouse model to test the efficiency and safety of concomitant treatment with IRI and THC. Male BALB/c mice subcutaneously injected with CT26 cells were receiving 60 mg/kg of IRI intraperitoneally on day 1 and 5 of treatment and/or 7 mg/kg of THC by gavage a day for 7 days. Treatment responses were evaluated based on changes in body, brain, and liver weight, tumour growth, blood cholinesterase activity, and oxidative stress parameters. Irinotecan's systemic toxicity was evidenced by weight loss and high oxidative stress. The important finding of this study is that combining THC with IRI diminishes IRI efficiency in inhibiting tumour growth. However, further studies, focused on more subtle molecular methods in tumour tissue and analytical analysis of IRI and THC distribution in tumour-bearing mice, are needed to prove our observations.
Collapse
Affiliation(s)
- Suzana Žunec
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | | | | | - Andreja Jurič
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Anja Katić
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Goran Kozina
- University North, University Centre Varaždin, Varaždin, Croatia
| | - Vedran Micek
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | | | | |
Collapse
|
7
|
El Omari N, Bakrim S, Khalid A, Albratty M, Abdalla AN, Lee LH, Goh KW, Ming LC, Bouyahya A. Anticancer clinical efficiency and stochastic mechanisms of belinostat. Biomed Pharmacother 2023; 165:115212. [PMID: 37541175 DOI: 10.1016/j.biopha.2023.115212] [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: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023] Open
Abstract
Cancer progression is strongly affected by epigenetic events in addition to genetic modifications. One of the key elements in the epigenetic control of gene expression is histone modification through acetylation, which is regulated by the synergy between histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDACs are thought to offer considerable potential for the development of anticancer medications, particularly when used in conjunction with other anticancer medications and/or radiotherapy. Belinostat (Beleodaq, PXD101) is a pan-HDAC unsaturated hydroxamate inhibitor with a sulfonamide group that has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of refractory or relapsed peripheral T-cell lymphoma (PTCL) and solid malignancies or and other hematological tissues. This drug modifies histones and epigenetic pathways. Because HDAC and HAT imbalance can lead to downregulation of regulatory genes, resulting in tumorigenesis. Inhibition of HDACs by belinostat indirectly promotes anti-cancer therapeutic effect by provoking acetylated histone accumulation, re-establishing normal gene expressions in cancer cells and stimulating other routes such as the immune response, p27 signaling cascades, caspase 3 activation, nuclear protein poly (ADP-ribose) polymerase-1 (PARP-1) degradation, cyclin A (G2/M phase), cyclin E1 (G1/S phase) and other events. In addition, belinostat has already been discovered to increase p21WAF1 in a number of cell lines (melanoma, prostate, breast, lung, colon, and ovary). This cyclin-dependent kinase inhibitor actually has a role in processes that cause cell cycle arrest and apoptosis. Belinostat's clinical effectiveness, comprising Phase I and II studies within the areas of solid and hematological cancers, has been evidenced through several investigative trials that have supported its potential to be a valuable anti-cancer drug. The purpose of this research was to provide insight on the specific molecular processes through which belinostat inhibits HDAC. The ability to investigate new therapeutic options employing targeted therapy and acquire a deeper understanding of cancer cell abnormalities may result from a better understanding of these particular routes.
Collapse
Affiliation(s)
- Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco; Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco; Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia; Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia; Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| | - Learn-Han Lee
- Sunway Microbiomics Centre, School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia; Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Malaysia.
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia.
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia.
| | - Abdelhakim Bouyahya
- School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia; Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco.
| |
Collapse
|
8
|
Gan C, Wang J, Wang Y, Martínez-Chávez A, Hillebrand M, de Vries N, Beukers J, Lebre MC, Wagenaar E, Rosing H, Klarenbeek S, Bleijerveld OB, Song JY, Altelaar M, Beijnen JH, Schinkel AH. Natural deletion of mouse carboxylesterases Ces1c/d/e impacts drug metabolism and metabolic syndrome development. Biomed Pharmacother 2023; 164:114956. [PMID: 37267638 DOI: 10.1016/j.biopha.2023.114956] [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: 03/03/2023] [Revised: 05/12/2023] [Accepted: 05/27/2023] [Indexed: 06/04/2023] Open
Abstract
Mammalian carboxylesterase 1 enzymes can hydrolyze many xenobiotic chemicals and endogenous lipids. We here identified and characterized a mouse strain (FVB/NKI) in which three of the eight Ces1 genes were spontaneously deleted, removing Ces1c and Ces1e partly, and Ces1d entirely. We studied the impact of this Ces1c/d/e deficiency on drug and lipid metabolism and homeostasis. Ces1c/d/e-/- mice showed strongly impaired conversion of the anticancer prodrug irinotecan to its active metabolite SN-38 in plasma, spleen and lung. Plasma hydrolysis of the oral anticancer prodrug capecitabine to 5-DFCR was also profoundly reduced in Ces1c/d/e-/- mice. Our findings resolved previously unexplained FVB/NKI pharmacokinetic anomalies. On a medium-fat diet, Ces1c/d/e-/- female mice exhibited moderately higher body weight, mild inflammation in gonadal white adipose tissue (gWAT), and increased lipid load in brown adipose tissue (BAT). Ces1c/d/e-/- males showed more pronounced inflammation in gWAT and an increased lipid load in BAT. On a 5-week high-fat diet exposure, Ces1c/d/e deficiency predisposed to developing obesity, enlarged and fatty liver, glucose intolerance and insulin resistance, with severe inflammation in gWAT and increased lipid load in BAT. Hepatic proteomics analysis revealed that the acute phase response, involved in the dynamic cycle of immunometabolism, was activated in these Ces1c/d/e-/- mice. This may contribute to the obesity-related chronic inflammation and adverse metabolic disease in this strain. While Ces1c/d/e deficiency clearly exacerbated metabolic syndrome development, long-term (18-week) high-fat diet exposure overwhelmed many, albeit not all, observed phenotypic differences.
Collapse
Affiliation(s)
- Changpei Gan
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Jing Wang
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Yaogeng Wang
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Alejandra Martínez-Chávez
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands; Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Michel Hillebrand
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Niels de Vries
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Joke Beukers
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Els Wagenaar
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Onno B Bleijerveld
- Proteomics Core Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Ji-Ying Song
- Experimental Animal Pathology Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - Maarten Altelaar
- Proteomics Core Facility, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, and Netherlands Proteomics Center, Padualaan 8, Utrecht 3584 CH, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands; Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CS, the Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands.
| |
Collapse
|
9
|
Miners JO, Polasek TM, Hulin JA, Rowland A, Meech R. Drug-drug interactions that alter the exposure of glucuronidated drugs: Scope, UDP-glucuronosyltransferase (UGT) enzyme selectivity, mechanisms (inhibition and induction), and clinical significance. Pharmacol Ther 2023:108459. [PMID: 37263383 DOI: 10.1016/j.pharmthera.2023.108459] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Drug-drug interactions (DDIs) arising from the perturbation of drug metabolising enzyme activities represent both a clinical problem and a potential economic loss for the pharmaceutical industry. DDIs involving glucuronidated drugs have historically attracted little attention and there is a perception that interactions are of minor clinical relevance. This review critically examines the scope and aetiology of DDIs that result in altered exposure of glucuronidated drugs. Interaction mechanisms, namely inhibition and induction of UDP-glucuronosyltransferase (UGT) enzymes and the potential interplay with drug transporters, are reviewed in detail, as is the clinical significance of known DDIs. Altered victim drug exposure arising from modulation of UGT enzyme activities is relatively common and, notably, the incidence and importance of UGT induction as a DDI mechanism is greater than generally believed. Numerous DDIs are clinically relevant, resulting in either loss of efficacy or an increased risk of adverse effects, necessitating dose individualisation. Several generalisations relating to the likelihood of DDIs can be drawn from the known substrate and inhibitor selectivities of UGT enzymes, highlighting the importance of comprehensive reaction phenotyping studies at an early stage of drug development. Further, rigorous assessment of the DDI liability of new chemical entities that undergo glucuronidation to a significant extent has been recommended recently by regulatory guidance. Although evidence-based approaches exist for the in vitro characterisation of UGT enzyme inhibition and induction, the availability of drugs considered appropriate for use as 'probe' substrates in clinical DDI studies is limited and this should be research priority.
Collapse
Affiliation(s)
- John O Miners
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Thomas M Polasek
- Certara, Princeton, NJ, USA; Centre for Medicines Use and Safety, Monash University, Melbourne, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Andrew Rowland
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
10
|
Obayashi N, Sakayori N, Kawaguchi H, Sugita M. Effect of irinotecan administration on amiloride-sensitive sodium taste responses in mice. Eur J Oral Sci 2023; 131:e12922. [PMID: 36852977 DOI: 10.1111/eos.12922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Taste alteration is a frequently reported side effect in patients receiving the chemotherapeutic agent, irinotecan. However, the way in which irinotecan causes taste disturbance and the type of taste impairment that is affected remain elusive. Here, we used the two-bottle preference test to characterize behavioral taste responses and employed immunohistochemical analyses to clarify the types and mechanisms of taste alteration induced, in mice, by irinotecan administration. Irinotecan administration resulted in a reduced intake of sodium taste solution but had no effect on sweet taste responses, as determined in the two-bottle preference test. In the presence of amiloride, which inhibits the function of the epithelial sodium channel (ENaC) in the periphery, the intake of sodium taste solution was comparable between the irinotecan-treated and control groups. Immunohistochemical analyses revealed that α-ENaC immunoreactivity detected in taste bud cells decreased slowly after irinotecan administration, and that administration of irinotecan had little effect on the number of cells expressing the cellular proliferation marker, Ki67, within or around taste buds. Our results imply that irinotecan administration may be responsible for altered behavioral sodium taste responses originating from ENaC function in the periphery, while being accompanied by the reduction of α-ENaC expression at the apical membrane of taste receptor cells without disturbing taste cell renewal.
Collapse
Affiliation(s)
- Nami Obayashi
- Department of Physiology and Oral Physiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of General Dentistry, Hiroshima University Hospital, Hiroshima, Japan
| | - Nobuyuki Sakayori
- Department of Physiology and Oral Physiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Kawaguchi
- Department of General Dentistry, Hiroshima University Hospital, Hiroshima, Japan
| | - Makoto Sugita
- Department of Physiology and Oral Physiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
11
|
Zhang Y, Wang J, Liu C, Xing H, Jiang Y, Li X. Novel disulfide bond bridged 7-ethyl-10-hydroxyl camptothecin-undecanoic acid conjugate/human serum albumin nanoparticles for breast cancer therapy. J Mater Chem B 2023; 11:2478-2489. [PMID: 36843543 DOI: 10.1039/d2tb02506j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
7-Ethyl-10-hydroxyl camptothecin (SN38), a semisynthetic derivative of camptothecin, exhibited extreme pharmacological activities in treating a range of cancers. However, its poor aqueous solubility and low stability hinder its clinical applications. Hence, a redox-responsive SN38 prodrug encapsulated human serum albumin (HSA) nanoparticle is developed to realize its potential in the clinic. First, a disulfide bond bridged 7-ethyl-10-hydroxyl camptothecin-undecanoic acid conjugate (SN38-SS-COOH) was synthesized and characterized structurally. After that, SN38-SS-COOH/HSA nanoparticles (SNH NPs) were prepared by the desolvation method. The SNH NPs with a feed molar ratio of 9 : 1 of SN38-SS-COOH : HSA showed a spherical structure with a diameter range of approximately 120-150 nm revealed by dynamic light scattering (DLS) and transmission electron microscopy (TEM). Fluorescence quenching confirmed the formation of SNH NP complexes by dual hydrophobic force and electrostatic interaction. The SNH NPs have a high drug loading of 10.44% and an encapsulation efficiency of 89.59% with good stability. Moreover, the redox responsiveness was validated by glutathione (GSH)-triggered accelerated release of parent drug SN38. In an in vivo pharmacokinetic study, the SNH NPs exhibited a significantly prolonged circulation time (t1/2, 3.77-fold) compared with free SN38. Finally, the in vivo antitumor efficacy and systemic toxicity of SNH NPs in a breast xenograft model were thoroughly evaluated. The inhibition rate of tumor growth induced by the SNH NPs reached 70.1%, while only 50.1% was achieved for irinotecan at an equivalent SN38 dosage of 10 mg kg-1. More importantly, the SNH NPs achieved a higher level of tumor growth inhibition (85.3%) by increasing the dosage to 60 mg kg-1 SN38 without obvious adverse effects. Taken together, the use of redox-responsive SN38 prodrug/HSA NPs could be a promising strategy to deliver highly active SN38 for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Yanhao Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Chao Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Hanlei Xing
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Yuhao Jiang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| |
Collapse
|
12
|
Aggarwal N, Kitano S, Puah GRY, Kittelmann S, Hwang IY, Chang MW. Microbiome and Human Health: Current Understanding, Engineering, and Enabling Technologies. Chem Rev 2023; 123:31-72. [PMID: 36317983 PMCID: PMC9837825 DOI: 10.1021/acs.chemrev.2c00431] [Citation(s) in RCA: 99] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Indexed: 01/12/2023]
Abstract
The human microbiome is composed of a collection of dynamic microbial communities that inhabit various anatomical locations in the body. Accordingly, the coevolution of the microbiome with the host has resulted in these communities playing a profound role in promoting human health. Consequently, perturbations in the human microbiome can cause or exacerbate several diseases. In this Review, we present our current understanding of the relationship between human health and disease development, focusing on the microbiomes found across the digestive, respiratory, urinary, and reproductive systems as well as the skin. We further discuss various strategies by which the composition and function of the human microbiome can be modulated to exert a therapeutic effect on the host. Finally, we examine technologies such as multiomics approaches and cellular reprogramming of microbes that can enable significant advancements in microbiome research and engineering.
Collapse
Affiliation(s)
- Nikhil Aggarwal
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Shohei Kitano
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Ginette Ru Ying Puah
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Wilmar-NUS
(WIL@NUS) Corporate Laboratory, National
University of Singapore, Singapore 117599, Singapore
- Wilmar
International Limited, Singapore 138568, Singapore
| | - Sandra Kittelmann
- Wilmar-NUS
(WIL@NUS) Corporate Laboratory, National
University of Singapore, Singapore 117599, Singapore
- Wilmar
International Limited, Singapore 138568, Singapore
| | - In Young Hwang
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Singapore
Institute of Technology, Singapore 138683, Singapore
| | - Matthew Wook Chang
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Wilmar-NUS
(WIL@NUS) Corporate Laboratory, National
University of Singapore, Singapore 117599, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| |
Collapse
|
13
|
Greco G, Zeppa SD, Agostini D, Attisani G, Stefanelli C, Ferrini F, Sestili P, Fimognari C. The Anti- and Pro-Tumorigenic Role of Microbiota and Its Role in Anticancer Therapeutic Strategies. Cancers (Basel) 2022; 15:190. [PMID: 36612186 PMCID: PMC9818275 DOI: 10.3390/cancers15010190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Human gut microbiota physiologically and actively participates as a symbiont to a wide number of fundamental biological processes, such as absorption and metabolism of nutrients, regulation of immune response and inflammation; gut microbiota plays also an antitumor role. However, dysbiosis, resulting from a number of different situations-dysmicrobism, infections, drug intake, age, diet-as well as from their multiple combinations, may lead to tumorigenesis and is associated with approximately 20% of all cancers. In a diagnostic, prognostic, therapeutic, and epidemiological perspective, it is clear that the bifaceted role of microbiota needs to be thoroughly studied and better understood. Here, we discuss the anti- and pro-tumorigenic potential of gut and other microbiota districts along with the causes that may change commensal bacteria from friend to foes.
Collapse
Affiliation(s)
- Giulia Greco
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, 40126 Bologna, Italy
| | - Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giuseppe Attisani
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy
| | - Claudio Stefanelli
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Carmela Fimognari
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy
| |
Collapse
|
14
|
D'Angelo A, Chapman R, Sirico M, Sobhani N, Catalano M, Mini E, Roviello G. An update on antibody-drug conjugates in urothelial carcinoma: state of the art strategies and what comes next. Cancer Chemother Pharmacol 2022; 90:191-205. [PMID: 35953604 PMCID: PMC9402760 DOI: 10.1007/s00280-022-04459-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022]
Abstract
In recent years, considerable progress has been made in increasing the knowledge of tumour biology and drug resistance mechanisms in urothelial cancer. Therapeutic strategies have significantly advanced with the introduction of novel approaches such as immune checkpoint inhibitors and Fibroblast Growth Factor Receptor inhibitors. However, despite these novel agents, advanced urothelial cancer is often still progressive in spite of treatment and correlates with a poor prognosis. The introduction of antibody–drug conjugates consisting of a target-specific monoclonal antibody covalently linked to a payload (cytotoxic agent) is a novel and promising therapeutic strategy. In December 2019, the US Food and Drug Administration (FDA) granted accelerated approval to the nectin-4-targeting antibody–drug conjugate, enfortumab vedotin, for the treatment of advanced or metastatic urothelial carcinomas that are refractory to both immune checkpoint inhibitors and platinum-based treatment. Heavily pre-treated urothelial cancer patients reported a significant, 40% response to enfortumab vedotin while other antibody–drug conjugates are currently still under investigation in several clinical trials. We have comprehensively reviewed the available treatment strategies for advanced urothelial carcinoma and outlined the mechanism of action of antibody–drug conjugate agents, their clinical applications, resistance mechanisms and future strategies for urothelial cancer.
Collapse
Affiliation(s)
- Alberto D'Angelo
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Robert Chapman
- Department of Medicine, Princess Alexandra Hospital NHS Foundation Trust, Harlow, CM20 1QX, UK
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Martina Catalano
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, vialePieraccini, 6, 50139, Florence, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, University of Florence, vialePieraccini, 6, 50139, Florence, Italy
| |
Collapse
|
15
|
Pant A, Maiti TK, Mahajan D, Das B. Human Gut Microbiota and Drug Metabolism. MICROBIAL ECOLOGY 2022:1-15. [PMID: 35869999 PMCID: PMC9308113 DOI: 10.1007/s00248-022-02081-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 07/18/2022] [Indexed: 05/31/2023]
Abstract
The efficacy of drugs widely varies in individuals, and the gut microbiota plays an important role in this variability. The commensal microbiota living in the human gut encodes several enzymes that chemically modify systemic and orally administered drugs, and such modifications can lead to activation, inactivation, toxification, altered stability, poor bioavailability, and rapid excretion. Our knowledge of the role of the human gut microbiome in therapeutic outcomes continues to evolve. Recent studies suggest the existence of complex interactions between microbial functions and therapeutic drugs across the human body. Therapeutic drugs or xenobiotics can influence the composition of the gut microbiome and the microbial encoded functions. Both these deviations can alter the chemical transformations of the drugs and hence treatment outcomes. In this review, we provide an overview of (i) the genetic ecology of microbially encoded functions linked with xenobiotic degradation; (ii) the effect of drugs on the composition and function of the gut microbiome; and (iii) the importance of the gut microbiota in drug metabolism.
Collapse
Affiliation(s)
- Archana Pant
- Molecular Genetics Lab, National Institute of Immunology, New Delhi, Delhi-110067, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad-121001, India
- Molecular Genetics Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, PO box, Gurgaon Expressway, #04 Faridabad-121001, Haryana, India
| | - Tushar K Maiti
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad-121001, India
| | - Dinesh Mahajan
- Chemistry and Pharmacology Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, PO box, Gurgaon Expressway, #04 Faridabad-121001, Haryana, India.
| |
Collapse
|
16
|
Xue T, Xu P, Padelford J, Xue X, Wu AY, Li Y, Wang L. Actively targeted delivery of SN38 by ultrafine iron oxide nanoparticle for treating pancreatic cancer. Invest New Drugs 2022; 40:546-555. [PMID: 35290548 DOI: 10.1007/s10637-022-01231-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/28/2022] [Indexed: 10/18/2022]
Abstract
Pancreatic cancer remains one of the most lethal cancers largely due to the inefficient delivery of therapeutics. Nanomaterials have been extensively investigated as drug delivery platforms, showing improved drug pharmacodynamics and pharmacokinetics. However, their applications in pancreatic cancer have not yet been successful due to limited tumor delivery caused by dense tumor stroma and distorted tumor vasculatures. Meanwhile, smaller-sized nanomaterials have shown improved tumor delivery and retention in various tumors, including pancreatic tumors, suggesting their potential in enhancing drug delivery. An ultrafine iron oxide nanoparticle (uIONP) was used to encapsulate 7-ethyl-10-hydroxyl camptothecin (SN38), the water-insoluble active metabolite of pancreatic cancer chemotherapy drug irinotecan. Insulin-like growth factor 1 (IGF-1) was conjugated to uIONP as a ligand for targeting pancreatic cancer cells overexpressing IGF-1 receptor (IGF1R). The SN38 loading and release profile were characterized. The pancreatic cancer cell targeting using IGF1-uIONP/SN38 and subsequently induced cell apoptosis were also investigated. IGF1-uIONP/SN38 demonstrated a stable drug loading in physiological pH with the loading efficiency of 68.2 ± 3.5% (SN38/Fe, wt%) and < 7% release for 24 h. In tumor-interstitial- and lysosomal-mimicking pH (6.5 and 5.5), 52.2 and 91.3% of encapsulated SN38 were released over 24 h. The IGF1-uIONP/SN38 exhibited specific receptor-mediated cell targeting and cytotoxicity Ato MiaPaCa-2 and Panc02 pancreatic cancer cells with IC50 of 11.8 ± 2.3 and 20.8 ± 3.5 nM, respectively, but not to HEK293 human embryonic kidney cells. IGF1-uIONP significantly improved the targeted SN38 delivery to pancreatic cancer cells, holding the potential for in vivo theranostic applications.
Collapse
Affiliation(s)
- Ting Xue
- Department of Radiology, Affiliated Longhua People's Hospital, the Third School of Clinical Medicine, Southern Medical University, Shenzhen, 518109, China
| | - Peijia Xu
- Department of Radiology, Affiliated Longhua People's Hospital, the Third School of Clinical Medicine, Southern Medical University, Shenzhen, 518109, China
| | | | - Xingkui Xue
- Department of Radiology, Affiliated Longhua People's Hospital, the Third School of Clinical Medicine, Southern Medical University, Shenzhen, 518109, China
| | - Alyssa Y Wu
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Yuancheng Li
- LLC, 5M Biomed, Atlanta, GA, 30333, USA.
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA.
| | - Liya Wang
- Department of Radiology, Affiliated Longhua People's Hospital, the Third School of Clinical Medicine, Southern Medical University, Shenzhen, 518109, China.
| |
Collapse
|
17
|
Samec T, Boulos J, Gilmore S, Hazelton A, Alexander-Bryant A. Peptide-based delivery of therapeutics in cancer treatment. Mater Today Bio 2022; 14:100248. [PMID: 35434595 PMCID: PMC9010702 DOI: 10.1016/j.mtbio.2022.100248] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
Current delivery strategies for cancer therapeutics commonly cause significant systemic side effects due to required high doses of therapeutic, inefficient cellular uptake of drug, and poor cell selectivity. Peptide-based delivery systems have shown the ability to alleviate these issues and can significantly enhance therapeutic loading, delivery, and cancer targetability. Peptide systems can be tailor-made for specific cancer applications. This review describes three peptide classes, targeting, cell penetrating, and fusogenic peptides, as stand-alone nanoparticle systems, conjugations to nanoparticle systems, or as the therapeutic modality. Peptide nanoparticle design, characteristics, and applications are discussed as well as peptide applications in the clinical space.
Collapse
Affiliation(s)
- Timothy Samec
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Jessica Boulos
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Serena Gilmore
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Anthony Hazelton
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Angela Alexander-Bryant
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| |
Collapse
|
18
|
Elz AS, Trevaskis NL, Porter CJH, Bowen JM, Prestidge CA. Smart design approaches for orally administered lipophilic prodrugs to promote lymphatic transport. J Control Release 2021; 341:676-701. [PMID: 34896450 DOI: 10.1016/j.jconrel.2021.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/22/2022]
Abstract
Challenges to effective delivery of drugs following oral administration has attracted growing interest over recent decades. Small molecule drugs (<1000 Da) are generally absorbed across the gastrointestinal tract into the portal blood and further transported to the systemic circulation via the liver. This can result in a significant reduction to the oral bioavailability of drugs that are metabolically labile and ultimately lead to ineffective exposure and treatment. Targeting drug delivery to the intestinal lymphatics is attracting increased attention as an alternative route of drug transportation providing multiple benefits. These include bypassing hepatic first-pass metabolism and selectively targeting disease reservoirs residing within the lymphatic system. The particular physicochemical requirements for drugs to be able to access the lymphatics after oral delivery include high lipophilicity (logP>5) and high long-chain triglyceride solubility (> 50 mg/g), properties required to enable drug association with the lipoprotein transport pathway. The majority of small molecule drugs, however, are not this lipophilic and therefore not substantially transported via the intestinal lymph. This has contributed to a growing body of investigation into prodrug approaches to deliver drugs to the lymphatic system by chemical manipulation. Optimised lipophilic prodrugs have the potential to increase lymphatic transport thereby improving oral pharmacokinetics via a reduction in first pass metabolism and may also target of disease-specific reservoirs within the lymphatics. This may provide advantages for current pharmacotherapy approaches for a wide array of pathological conditions, e.g. immune disease, cancer and metabolic disease, and also presents a promising approach for advanced vaccination strategies. In this review, specific emphasis is placed on medicinal chemistry strategies that have been successfully employed to design lipophilic prodrugs to deliberately enable lymphatic transport. Recent progress and opportunities in medicinal chemistry and drug delivery that enable new platforms for efficacious and safe delivery of drugs are critically evaluated.
Collapse
Affiliation(s)
- Aurelia S Elz
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Christopher J H Porter
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Joanne M Bowen
- School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
19
|
Kim JH, Vinh LB, Hur M, Koo SC, Park WT, Moon YH, Lee YJ, Kim YH, Huh YC, Yang SY. Inhibitory Activity of 4- O-Benzoyl-3'- O-(OMethylsinapoyl) Sucrose from Polygala tenuifolia on Escherichia coliβ-Glucuronidase. J Microbiol Biotechnol 2021; 31:1576-1582. [PMID: 34528918 PMCID: PMC9705844 DOI: 10.4014/jmb.2108.08004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/15/2022]
Abstract
Bacterial β-glucuronidase in the intestine is involved in the conversion of 7-ethyl-10- hydroxycamptochecin glucuronide (derived from irinotecan) to 7-ethyl-10-hydroxycamptothecin, which causes intestinal bleeding and diarrhea (side effects of anti-cancer drugs). Twelve compounds (1-12) from Polygala tenuifolia were evaluated in terms of β-glucuronidase inhibition in vitro. 4-O-Benzoyl-3'-O-(O-methylsinapoyl) sucrose (C3) was highly inhibitory at low concentrations. C3 (an uncompetitive inhibitor) exhibited a ki value of 13.4 μM; inhibitory activity increased as the substrate concentration rose. Molecular simulation revealed that C3 bound principally to the Gln158-Tyr160 enzyme loop. Thus, C3 will serve as a lead compound for development of new β- glucuronidase inhibitors.
Collapse
Affiliation(s)
- Jang Hoon Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Le Ba Vinh
- Institute of Marine Biochemistry(IMBC), Vietnam Academy of Science and Technology(VAST), Hanoi 100000, Vietnam
| | - Mok Hur
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Sung-Cheol Koo
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Woo Tae Park
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Youn-Ho Moon
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Yoon Jeong Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Young Ho Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yun-Chan Huh
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea,
Y.C. Huh Phone: +82-43-871-5662 Fax: +82-43-871-5659 E-mail:
| | - Seo Young Yang
- Department of Pharmaceutical Engineering, Sangji University, Wonju 26339, Republic of Korea,Corresponding authors S.Y. Yang Phone: +82-33-738-7921 Fax: +82-33-738-7652 E-mail:
| |
Collapse
|
20
|
Hesse J, Martinelli J, Aboumanify O, Ballesta A, Relógio A. A mathematical model of the circadian clock and drug pharmacology to optimize irinotecan administration timing in colorectal cancer. Comput Struct Biotechnol J 2021; 19:5170-5183. [PMID: 34630937 PMCID: PMC8477139 DOI: 10.1016/j.csbj.2021.08.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/21/2022] Open
Abstract
Scheduling anticancer drug administration over 24 h may critically impact treatment success in a patient-specific manner. Here, we address personalization of treatment timing using a novel mathematical model of irinotecan cellular pharmacokinetics and -dynamics linked to a representation of the core clock and predict treatment toxicity in a colorectal cancer (CRC) cellular model. The mathematical model is fitted to three different scenarios: mouse liver, where the drug metabolism mainly occurs, and two human colorectal cancer cell lines representing an in vitro experimental system for human colorectal cancer progression. Our model successfully recapitulates quantitative circadian datasets of mRNA and protein expression together with timing-dependent irinotecan cytotoxicity data. The model also discriminates time-dependent toxicity between the different cells, suggesting that treatment can be optimized according to their cellular clock. Our results show that the time-dependent degradation of the protein mediating irinotecan activation, as well as an oscillation in the death rate may play an important role in the circadian variations of drug toxicity. In the future, this model can be used to support personalized treatment scheduling by predicting optimal drug timing based on the patient's gene expression profile.
Collapse
Affiliation(s)
- Janina Hesse
- Institute for Systems Medicine, Department of Human Medicine, MSH Medical School Hamburg - University of Applied Sciences and Medical University, Hamburg 20457, Germany.,Institute for Theoretical Biology (ITB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany
| | - Julien Martinelli
- INSERM U900, Saint-Cloud, France, Institut Curie, Saint Cloud, France, Paris Saclay University, France, MINES ParisTech, CBIO - Centre for Computational Biology, PSL Research University, Paris, France.,UPR 'Chronotherapy, Cancers and Transplantation', Faculty of Medicine, Paris Saclay University, Campus CNRS, 7 rue Guy Moquet, 94800 Villejuif, France.,Lifeware Group, Inria Saclay Ile-de-France, Palaiseau 91120, France
| | - Ouda Aboumanify
- Institute for Theoretical Biology (ITB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany.,Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin
| | - Annabelle Ballesta
- INSERM U900, Saint-Cloud, France, Institut Curie, Saint Cloud, France, Paris Saclay University, France, MINES ParisTech, CBIO - Centre for Computational Biology, PSL Research University, Paris, France.,UPR 'Chronotherapy, Cancers and Transplantation', Faculty of Medicine, Paris Saclay University, Campus CNRS, 7 rue Guy Moquet, 94800 Villejuif, France
| | - Angela Relógio
- Institute for Systems Medicine, Department of Human Medicine, MSH Medical School Hamburg - University of Applied Sciences and Medical University, Hamburg 20457, Germany.,Institute for Theoretical Biology (ITB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Berlin 10117, Germany.,Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin
| |
Collapse
|
21
|
Chi LH, Burrows AD, Anderson RL. Can preclinical drug development help to predict adverse events in clinical trials? Drug Discov Today 2021; 27:257-268. [PMID: 34469805 DOI: 10.1016/j.drudis.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/03/2021] [Accepted: 08/24/2021] [Indexed: 12/23/2022]
Abstract
The development of novel therapeutics is associated with high rates of attrition, with unexpected adverse events being a major cause of failure. Serious adverse events have led to organ failure, cancer development and deaths that were not expected outcomes in clinical trials. These life-threatening events were not identified during therapeutic development due to the lack of preclinical safety tests that faithfully represented human physiology. We highlight the successful application of several novel technologies, including high-throughput screening, organs-on-chips, microbiome-containing drug-testing platforms and humanised mouse models, for mechanistic studies and prediction of toxicity. We propose the incorporation of similar preclinical tests into future drug development to reduce the likelihood of hazardous therapeutics entering later-stage clinical trials.
Collapse
Affiliation(s)
- Lap Hing Chi
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Allan D Burrows
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Robin L Anderson
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
22
|
Samare-Najaf M, Samareh A, Jamali N, Abbasi A, Clark CC, Khorchani MJ, Zal F. Adverse Effects and Safety of Etirinotecan Pegol, a Novel Topoisomerase Inhibitor, in Cancer Treatment: A Systematic Review. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394717666210202103502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Due to the increasing prevalence of cancer and the inadequacy of current
therapies, the development of novel antitumor pharmaceutics with higher efficacies and lower adverse
effects is considered a fundamental tenet of contemporary cancer management.
Poly-Ethylene-Glycol (PEG) attachment is a novel pharmaceutical technology to improve the efficacy
and safety of chemotherapies. Etirinotecan Pegol (EP), also known as NKTR-102, is the PEGylated
form of Irinotecan (CPT-11), which causes cancer cell apoptosis by inhibiting the
topoisomerase I enzyme.
Objectives:
The present study reviews and evaluates various reports of the EP’s anti-tumor activity
in various cancers.
Data Sources:
Studies were identified using the Scopus database, with no exclusions. The search
terms included Etirinotecan Pegol and NKTR-102, which yielded 125 articles (66 and 59 articles,
respectively). In addition, the clinicaltrials.gov website was used to find ongoing studies, which resulted
in the addition of two studies.
Study Eligibility Criteria:
Subsequently, we excluded studies that were published in languages
other than English, duplicate articles, and studies with no data.
Results:
This systematic review clarifies that EP possesses numerous advantages over many other
medications, such as safety, efficacy, increased half-life, increased health-related quality of life, increased
overall survival, increased progression-free survival, and decreasing the adverse events in
the treatment of various cancers.
Conclusion:
Therefore, Etirinotecan Pegol may represent a major contribution to the treatment of
various cancers in the future.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Navid Jamali
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Abbasi
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cain C.T. Clark
- Centre for Intelligent Healthcare, Coventry University, CV1 5FB, United Kingdom
| | - Majid J. Khorchani
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Zal
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
23
|
Wang Y, Xie H, Ying K, Xie B, Chen X, Yang B, Jin J, Wan J, Li T, Han W, Fang S, Wang H. Tuning the efficacy of esterase-activatable prodrug nanoparticles for the treatment of colorectal malignancies. Biomaterials 2021; 270:120705. [PMID: 33581609 DOI: 10.1016/j.biomaterials.2021.120705] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/23/2021] [Accepted: 01/30/2021] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) is one of the most common and lethal human cancers, and the clinical outcomes remain unsatisfactory because of the lack of effective and safe therapeutic regimens. Here, we describe a practical and potent delivery approach for the human topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN38) against CRC. Injectable SN38-loaded nanoparticles are obtained through covalent ligation of the SN38 agent with oligo-ε-caprolactone (oligoCL) to form oligoCL-SN38 conjugates via an esterase-activatable linkage followed by encapsulation of these prodrugs in exogenous polymer matrices. Prodrug nanoparticles with adaptive features are sufficiently stable during blood circulation, while active drugs can be released in response to intracellular esterase. The administration of nanoparticle drugs results in durable tumor recession, and the efficacy is superior to that of the current standard-of-care therapy, CPT-11, in multiple mouse models of CRC, one of which is a chemically induced orthotopic CRC. Elucidation of the mechanism underlying these differing efficacies shows that nanoparticle delivery produces a substantial increase in the intratumoral concentration of the therapeutic agent relative to CPT-11, which contributes to improved antitumor efficacy. Finally, these nanoparticle drugs are potentially less toxic in animals than CPT-11, as evidenced by the low incidence of bloody diarrhea and attenuated colonic damage. Overall, these results demonstrate that precisely engineered therapeutic nanoparticles are capable of enhancing efficacy, addressing the risk of tumor recurrence, and increasing drug tolerance, thus deserving further investigation.
Collapse
Affiliation(s)
- Yuchen Wang
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China; Department of Chemical Engineering, Zhejiang University, Hangzhou, Zhejiang Province, 310027, PR China
| | - Haiyang Xie
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China
| | - Kangkang Ying
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310016, PR China
| | - Binbin Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310016, PR China
| | - Xiaona Chen
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China
| | - Bing Yang
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China
| | - Jiahui Jin
- Xingzhi College, Zhejiang Normal University, Jinhua, Zhejiang Province, 321004, PR China
| | - Jianqin Wan
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China
| | - Tongyu Li
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310016, PR China
| | - Shijiang Fang
- Department of Chemical Engineering, Zhejiang University, Hangzhou, Zhejiang Province, 310027, PR China
| | - Hangxiang Wang
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, PR China.
| |
Collapse
|
24
|
Bechtold B, Clarke J. Multi-factorial pharmacokinetic interactions: unraveling complexities in precision drug therapy. Expert Opin Drug Metab Toxicol 2020; 17:397-412. [PMID: 33339463 DOI: 10.1080/17425255.2021.1867105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Precision drug therapy requires accounting for pertinent factors in pharmacokinetic (PK) inter-individual variability (i.e., pharmacogenetics, diseases, polypharmacy, and natural product use) that can cause sub-therapeutic or adverse effects. Although each of these individual factors can alter victim drug PK, multi-factorial interactions can cause additive, synergistic, or opposing effects. Determining the magnitude and direction of these complex multi-factorial effects requires understanding the rate-limiting redundant and/or sequential PK processes for each drug.Areas covered: Perturbations in drug-metabolizing enzymes and/or transporters are integral to single- and multi-factorial PK interactions. Examples of single factor PK interactions presented include gene-drug (pharmacogenetic), disease-drug, drug-drug, and natural product-drug interactions. Examples of multi-factorial PK interactions presented include drug-gene-drug, natural product-gene-drug, gene-gene-drug, disease-natural product-drug, and disease-gene-drug interactions. Clear interpretation of multi-factorial interactions can be complicated by study design, complexity in victim drug PK, and incomplete mechanistic understanding of victim drug PK.Expert opinion: Incorporation of complex multi-factorial PK interactions into precision drug therapy requires advances in clinical decision tools, intentional PK study designs, drug-metabolizing enzyme and transporter fractional contribution determinations, systems and computational approaches (e.g., physiologically-based pharmacokinetic modeling), and PK phenotyping of progressive diseases.
Collapse
Affiliation(s)
- Baron Bechtold
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - John Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| |
Collapse
|
25
|
Fidelle M, Yonekura S, Picard M, Cogdill A, Hollebecque A, Roberti MP, Zitvogel L. Resolving the Paradox of Colon Cancer Through the Integration of Genetics, Immunology, and the Microbiota. Front Immunol 2020; 11:600886. [PMID: 33381121 PMCID: PMC7768083 DOI: 10.3389/fimmu.2020.600886] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
While colorectal cancers (CRC) are paradigmatic tumors invaded by effector memory lymphocytes, the mechanisms accounting for the relative resistance of MSI negative CRC to immunogenic cell death mediated by oxaliplatin and immune checkpoint inhibitors has remained an open conundrum. Here, we propose the viewpoint where its microenvironmental contexture could be explained -at least in part- by macroenvironmental cues constituted by the complex interplay between the epithelial barrier, its microbial ecosystem, and the local immune system. Taken together this dynamic ménage-à-trois offers novel coordinated actors of the humoral and cellular immune responses actionable to restore sensitivity to immune checkpoint inhibition. Solving this paradox involves breaking tolerance to crypt stem cells by inducing the immunogenic apoptosis of ileal cells in the context of an ileal microbiome shifted towards immunogenic bacteria using cytotoxicants. This manoeuver results in the elicitation of a productive Tfh and B cell dialogue in mesenteric lymph nodes culminating in tumor-specific memory CD8+ T cell responses sparing the normal epithelium.
Collapse
Affiliation(s)
- Marine Fidelle
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Satoru Yonekura
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Marion Picard
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Unit Biology and Genetics of the Bacterial Cell Wall, Institut Pasteur, Paris, France
| | - Alexandria Cogdill
- Department of Immunology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
- Department of Genomic Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Antoine Hollebecque
- Gustave Roussy, Villejuif, France
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Villejuif, France
- Equipe Labellisée—Ligue Nationale contre le Cancer, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
| |
Collapse
|
26
|
Yamasaki K, Hidaka M, Kawano Y, Furuya Y, Ono H, Arimori K. Possible roles of intestinal P-glycoprotein and cytochrome P450 3A on the limited oral absorption of irinotecan. J Pharm Pharmacol 2020; 73:178-184. [PMID: 33793796 DOI: 10.1093/jpp/rgaa009] [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: 06/26/2020] [Accepted: 09/30/2020] [Indexed: 11/12/2022]
Abstract
OBJECTIVES Irinotecan is a widely intravenously used drug for the treatment of certain types of solid tumours. The oral administration of irinotecan has recently been recognized as being a more effective method for the treatment than intravenous administration. However, the limited oral bioavailability of irinotecan poses a problem for its oral delivery. In this study, we report on an investigation of the mechanism responsible for the limited oral absorption of irinotecan using rats as models. METHODS The intestinal absorption of irinotecan in the absence and presence of several compounds was examined using intestinal loop method. The pharmacokinetics of irinotecan was investigated when verapamil, an inhibitor of the P-glycoprotein (P-gp) and cytochrome P450 3A (CYP3A) was pre-administered. KEY FINDINGS The intestinal absorption of irinotecan was enhanced in the presence of verapamil, indicating that efflux by intestinal P-gp contributes to its limited oral absorption. Indeed, the oral bioavailability of irinotecan was increased when verapamil was orally pre-administered. This increased oral bioavailability was accompanied by a slight but significant decrease in the formation of a metabolite produced by the action of CYP3A. CONCLUSION The findings presented herein suggest that intestinal efflux by P-gp is mainly and intestinal metabolism by CYP3A is partially responsible for the limited oral absorption of irinotecan.
Collapse
Affiliation(s)
- Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.,DDS Research Institute, Sojo University, Kumamoto, Japan.,Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan
| | - Muneaki Hidaka
- Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan.,Department of Clinical Pharmacy, Graduate School of Clinical Pharmacy, Kyushu University of Health and Welfare, Nobeoka, Japan
| | - Yohei Kawano
- Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan.,Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yumiko Furuya
- Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan
| | - Hiroshige Ono
- Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan.,Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda, Japan
| | - Kazuhiko Arimori
- Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan
| |
Collapse
|
27
|
Matsunaga T, Okumura N, Saito H, Morikawa Y, Suenami K, Hisamatsu A, Endo S, Ikari A. Significance of aldo-keto reductase 1C3 and ATP-binding cassette transporter B1 in gain of irinotecan resistance in colon cancer cells. Chem Biol Interact 2020; 332:109295. [PMID: 33096057 DOI: 10.1016/j.cbi.2020.109295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 11/29/2022]
Abstract
Irinotecan (CPT11) is widely prescribed for treatment of various intractable cancers such as advanced and metastatic colon cancer cells, but its continuous treatment promotes the resistance development. In this study, we established CPT11-resistant variants of three human colon cancer (DLD1, RKO and LoVo) cell lines, and found that gain of the resistance elicited an up-regulation of aldo-keto reductase (AKR) 1C3 in the cells. Additionally, the sensitivity to CPT11 toxicity was decreased and increased by overexpression and knockdown, respectively, of the enzyme. Moreover, the resistant cells suppressed formation of reactive 4-hydroxy-2-nonenal by CPT11 treatment, and the suppressive effect was almost completely abolished by addition of an AKR1C3 inhibitor. These results suggest that up-regulated AKR1C3 contributes to promotion of the chemoresistance by detoxifying the reactive aldehyde. Western blot and real-time polymerase-chain reaction analyses and ATP-binding cassette (ABC) B1-functional assay revealed that, among three ABC transporters, ABCB1 was the most highly up-regulated by development of the CPT11 resistance, inferring a significant contribution of pregnane-X receptor-dependent signaling to the ABCB1 up-regulation. The combined treatment with inhibitors of AKR1C3 and ABCB1 potently sensitized the resistant cells to CPT11 and its active metabolite SN38. Taken together, our results suggest that combination of AKR1C3 and ABCB1 inhibitors is effective as adjuvant therapy to enhance CPT11 sensitivity of intractable colon cancer cells.
Collapse
Affiliation(s)
- Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 502-8585, Japan.
| | - Naoko Okumura
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Haruhi Saito
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Koichi Suenami
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Aki Hisamatsu
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| |
Collapse
|
28
|
Effect of drug metabolizing enzymes and transporters in Thai colorectal cancer patients treated with irinotecan-based chemotherapy. Sci Rep 2020; 10:13486. [PMID: 32778670 PMCID: PMC7417535 DOI: 10.1038/s41598-020-70351-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 07/21/2020] [Indexed: 12/30/2022] Open
Abstract
Genetic polymorphisms in drug metabolizing enzymes and drug transporters may affect irinotecan toxicity. Although genetic polymorphisms have been shown to influence the irinotecan toxicity, data are limited in Thai population. Thus, the aim of this study was to assess the allele and genotype frequencies and the relationship between CYP3A4/5, DPYD, UGT1A1, ABCB1, and ABCC2 genetic variations and irinotecan-induced toxicity in Thai colorectal cancer patients. One hundred and thirty-two patients were genotyped, and the effect of genetic variations on irinotecan-induced toxicity was assessed in 66 patients who received irinotecan-based chemotherapy. Allele frequencies of ABCB1 c.1236C > T, ABCB1 c.3435C > T, ABCC2 c.3972C > T, ABCG2 c.421C > A, CYP3A4*1B, CYP3A4*18, CYP3A5*3, DPYD*5, UGT1A1*28, and UGT1A1*6 were 0.67, 0.43, 0.23, 0.27, 0.01, 0.02, 0.64, 0.19, 0.16, and 0.09, respectively. DPYD*2A and DPYD c.1774C > T variants were not detected in our study population. The ABCC2 c.3972C > T was significantly associated with grade 1–4 neutropenia (P < 0.012) at the first cycle. Patients carrying both UGT1A1*28 and *6 were significantly associated with severe neutropenia at the first (P < 0.001) and second (P = 0.017) cycles. In addition, patients carrying UG1A1*28 and *6 had significantly lower absolute neutrophil count (ANC) nadir at first (P < 0.001) and second (P = 0.001) cycles. This finding suggests that UGT1A1*28, *6, and ABCC2 c.3972C > T might be an important predictor for irinotecan-induced severe neutropenia.
Collapse
|
29
|
Kopjar N, Fuchs N, Brčić Karačonji I, Žunec S, Katić A, Kozina G, Lucić Vrdoljak A. High Doses of Δ 9-Tetrahydrocannabinol Might Impair Irinotecan Chemotherapy: A Review of Potentially Harmful Interactions. Clin Drug Investig 2020; 40:775-787. [PMID: 32696321 DOI: 10.1007/s40261-020-00954-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review proposes the hypothesis that the effectiveness of irinotecan chemotherapy might be impaired by high doses of concomitantly administered Δ9-tetrahydrocannabinol (THC). The most important features shared by irinotecan and THC, which might represent sources of potentially harmful interactions are: first-pass hepatic metabolism mediated by cytochrome P450 (CYP) enzyme CYP3A4; glucuronidation mediated by uridine diphosphate glycosyltransferase (UGT) enzymes, isoforms 1A1 and 1A9; transport of parent compounds and their metabolites via canalicular ATP-binding cassette (ABC) transporters ABCB1 and ABCG2; enterohepatic recirculation of both parent compounds, which leads to an extended duration of their pharmacological effects; possible competition for binding to albumin; butyrylcholinesterase (BChE) inhibition by THC, which might impair the conversion of parent irinotecan into the SN-38 metabolite; mutual effects on mitochondrial dysfunction and induction of oxidative stress; potentiation of hepatotoxicity; potentiation of genotoxicity and cytogenetic effects leading to genome instability; possible neurotoxicity; and effects on bilirubin. The controversies associated with the use of highly concentrated THC preparations with irinotecan chemotherapy are also discussed. Despite all of the limitations, the body of evidence provided here could be considered relevant for human-risk assessments and calls for concern in cases when irinotecan chemotherapy is accompanied by preparations rich in THC.
Collapse
Affiliation(s)
- Nevenka Kopjar
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Nino Fuchs
- Department of Surgery, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Irena Brčić Karačonji
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Suzana Žunec
- Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Anja Katić
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Goran Kozina
- University Centre Varaždin, University North, Varaždin, Croatia
| | - Ana Lucić Vrdoljak
- Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| |
Collapse
|
30
|
Irinotecan-Still an Important Player in Cancer Chemotherapy: A Comprehensive Overview. Int J Mol Sci 2020; 21:ijms21144919. [PMID: 32664667 PMCID: PMC7404108 DOI: 10.3390/ijms21144919] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 02/06/2023] Open
Abstract
Irinotecan has been used in the treatment of various malignancies for many years. Still, the knowledge regarding this drug is expanding. The pharmacogenetics of the drug is the crucial component of response to irinotecan. Furthermore, new formulations of the drug are introduced in order to better deliver the drug and avoid potentially life-threatening side effects. Here, we give a comprehensive overview on irinotecan’s molecular mode of action, metabolism, pharmacogenetics, and toxicity. Moreover, this article features clinically used combinations of the drug with other anticancer agents and introduces novel formulations of drugs (e.g., liposomal formulations, dendrimers, and nanoparticles). It also outlines crucial mechanisms of tumor cells’ resistance to the active metabolite, ethyl-10-hydroxy-camptothecin (SN-38). We are sure that the article will constitute an important source of information for both new researchers in the field of irinotecan chemotherapy and professionals or clinicians who are interested in the topic.
Collapse
|
31
|
Characterization of the Blood-Brain Barrier Integrity and the Brain Transport of SN-38 in an Orthotopic Xenograft Rat Model of Diffuse Intrinsic Pontine Glioma. Pharmaceutics 2020; 12:pharmaceutics12050399. [PMID: 32349240 PMCID: PMC7284501 DOI: 10.3390/pharmaceutics12050399] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/20/2020] [Accepted: 04/25/2020] [Indexed: 11/17/2022] Open
Abstract
The blood-brain barrier (BBB) hinders the brain delivery of many anticancer drugs. In pediatric patients, diffuse intrinsic pontine glioma (DIPG) represents the main cause of brain cancer mortality lacking effective drug therapy. Using sham and DIPG-bearing rats, we analyzed 1) the brain distribution of 3-kDa-Texas red-dextran (TRD) or [14C]-sucrose as measures of BBB integrity, and 2) the role of major ATP-binding cassette (ABC) transporters at the BBB on the efflux of the irinotecan metabolite [3H]-SN-38. The unaffected [14C]-sucrose or TRD distribution in the cerebrum, cerebellum, and brainstem regions in DIPG-bearing animals suggests an intact BBB. Targeted proteomics retrieved no change in P-glycoprotein (P-gp), BCRP, MRP1, and MRP4 levels in the analyzed regions of DIPG rats. In vitro, DIPG cells express BCRP but not P-gp, MRP1, or MRP4. Dual inhibition of P-gp/Bcrp, or Mrp showed a significant increase on SN-38 BBB transport: Cerebrum (8.3-fold and 3-fold, respectively), cerebellum (4.2-fold and 2.8-fold), and brainstem (2.6-fold and 2.2-fold). Elacridar increased [3H]-SN-38 brain delivery beyond a P-gp/Bcrp inhibitor effect alone, emphasizing the role of another unidentified transporter in BBB efflux of SN-38. These results confirm a well-preserved BBB in DIPG-bearing rats, along with functional ABC-transporter expression. The development of chemotherapeutic strategies to circumvent ABC-mediated BBB efflux are needed to improve anticancer drug delivery against DIPG.
Collapse
|
32
|
Bhatt AP, Pellock SJ, Biernat KA, Walton WG, Wallace BD, Creekmore BC, Letertre MM, Swann JR, Wilson ID, Roques JR, Darr DB, Bailey ST, Montgomery SA, Roach JM, Azcarate-Peril MA, Sartor RB, Gharaibeh RZ, Bultman SJ, Redinbo MR. Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy. Proc Natl Acad Sci U S A 2020; 117:7374-7381. [PMID: 32170007 PMCID: PMC7132129 DOI: 10.1073/pnas.1918095117] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Irinotecan treats a range of solid tumors, but its effectiveness is severely limited by gastrointestinal (GI) tract toxicity caused by gut bacterial β-glucuronidase (GUS) enzymes. Targeted bacterial GUS inhibitors have been shown to partially alleviate irinotecan-induced GI tract damage and resultant diarrhea in mice. Here, we unravel the mechanistic basis for GI protection by gut microbial GUS inhibitors using in vivo models. We use in vitro, in fimo, and in vivo models to determine whether GUS inhibition alters the anticancer efficacy of irinotecan. We demonstrate that a single dose of irinotecan increases GI bacterial GUS activity in 1 d and reduces intestinal epithelial cell proliferation in 5 d, both blocked by a single dose of a GUS inhibitor. In a tumor xenograft model, GUS inhibition prevents intestinal toxicity and maintains the antitumor efficacy of irinotecan. Remarkably, GUS inhibitor also effectively blocks the striking irinotecan-induced bloom of Enterobacteriaceae in immune-deficient mice. In a genetically engineered mouse model of cancer, GUS inhibition alleviates gut damage, improves survival, and does not alter gut microbial composition; however, by allowing dose intensification, it dramatically improves irinotecan's effectiveness, reducing tumors to a fraction of that achieved by irinotecan alone, while simultaneously promoting epithelial regeneration. These results indicate that targeted gut microbial enzyme inhibitors can improve cancer chemotherapeutic outcomes by protecting the gut epithelium from microbial dysbiosis and proliferative crypt damage.
Collapse
Affiliation(s)
- Aadra P Bhatt
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
| | - Samuel J Pellock
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
| | - Kristen A Biernat
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
| | - William G Walton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
| | - Bret D Wallace
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
| | - Benjamin C Creekmore
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
| | - Marine M Letertre
- Computational and Systems Medicine, Department of Surgery & Cancer, Imperial College London, SW7 2AZ London, United Kingdom
| | - Jonathan R Swann
- Computational and Systems Medicine, Department of Surgery & Cancer, Imperial College London, SW7 2AZ London, United Kingdom
| | - Ian D Wilson
- Computational and Systems Medicine, Department of Surgery & Cancer, Imperial College London, SW7 2AZ London, United Kingdom
| | - Jose R Roques
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - David B Darr
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Sean T Bailey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Stephanie A Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7525
| | - Jeffrey M Roach
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
| | - R Balfour Sartor
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7555
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Raad Z Gharaibeh
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610
| | - Scott J Bultman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7264
| | - Matthew R Redinbo
- Department of Biochemistry, Integrated Program for Biological and Genome Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290;
- Department of Biophysics, Integrated Program for Biological and Genome Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3290
| |
Collapse
|
33
|
Abstract
The intestinal microbiome encodes vast metabolic potential, and multidisciplinary approaches are enabling a mechanistic understanding of how bacterial enzymes impact the metabolism of diverse pharmaceutical compounds, including chemotherapeutics. Microbiota alter the activity of many drugs and chemotherapeutics via direct and indirect mechanisms; some of these alterations result in changes to the drug's bioactivity and bioavailability, causing toxic gastrointestinal side effects. Gastrointestinal toxicity is one of the leading complications of systemic chemotherapy, with symptoms including nausea, vomiting, diarrhea, and constipation. Patients undergo dose reductions or drug holidays to manage these adverse events, which can significantly harm prognosis, and can result in mortality. Selective and precise targeting of the gut microbiota may alleviate these toxicities. Understanding the composition and function of the microbiota may serve as a biomarker for prognosis, and predict treatment efficacy and potential adverse effects, thereby facilitating personalized medicine strategies for cancer patients.
Collapse
Affiliation(s)
- Samantha M. Ervin
- Department of Chemistry, University of North Carolina at Chapel Hill, 250 Bell Tower Drive, Chapel Hill, NC 27599, USA
| | | | - Aadra P. Bhatt
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, USA.,Corresponding author:
| |
Collapse
|
34
|
Collins SL, Patterson AD. The gut microbiome: an orchestrator of xenobiotic metabolism. Acta Pharm Sin B 2020; 10:19-32. [PMID: 31998605 PMCID: PMC6984741 DOI: 10.1016/j.apsb.2019.12.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/21/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Microbes inhabiting the intestinal tract of humans represent a site for xenobiotic metabolism. The gut microbiome, the collection of microorganisms in the gastrointestinal tract, can alter the metabolic outcome of pharmaceuticals, environmental toxicants, and heavy metals, thereby changing their pharmacokinetics. Direct chemical modification of xenobiotics by the gut microbiome, either through the intestinal tract or re-entering the gut via enterohepatic circulation, can lead to increased metabolism or bioactivation, depending on the enzymatic activity within the microbial niche. Unique enzymes encoded within the microbiome include those that reverse the modifications imparted by host detoxification pathways. Additionally, the microbiome can limit xenobiotic absorption in the small intestine by increasing the expression of cell-cell adhesion proteins, supporting the protective mucosal layer, and/or directly sequestering chemicals. Lastly, host gene expression is regulated by the microbiome, including CYP450s, multi-drug resistance proteins, and the transcription factors that regulate them. While the microbiome affects the host and pharmacokinetics of the xenobiotic, xenobiotics can also influence the viability and metabolism of the microbiome. Our understanding of the complex interconnectedness between host, microbiome, and metabolism will advance with new modeling systems, technology development and refinement, and mechanistic studies focused on the contribution of human and microbial metabolism.
Collapse
Key Words
- 5-ASA, 5-aminosalicylic acid
- 5-FU, 5-fluorouracil
- AHR, aryl Hydrocarbon Receptor
- ALDH, aldehyde dehydrogenase
- Absorption
- BDE, bromodiphenyl ether
- BRV, brivudine
- BVU, bromovinyluracil
- Bioactivation
- CAR, constitutive androgen receptor
- CV, conventional
- CYP, cytochrome P450
- ER, estrogen receptor
- Enterohepatic circulation
- FXR, farnesoid X receptor
- GF, germ-free
- GUDCA, glycoursodeoxycholic acid
- Gastrointestinal tract
- Gut microbiome
- NSAID, non-steroidal anti-inflammatory drug
- PABA, p-aminobenzenesulphonamide
- PAH, polycyclic aromatic hydrocarbon
- PCB, polychlorinated biphenyl
- PD, Parkinson's disease
- PFOS, perfluorooctanesulfonic acid
- PXR, pregnane X receptor
- Pharmacokinetics
- SCFA, short chain fatty acid
- SN-38G, SN-38 glucuronide
- SULT, sulfotransferase
- TCDF, 2,3,7,8-tetrachlorodibenzofuran
- TUDCA, tauroursodeoxycholic acid
- UGT, uracil diphosphate-glucuronosyltransferase
- Xenobiotic metabolism
- cgr, cytochrome glycoside reductase
Collapse
Affiliation(s)
- Stephanie L. Collins
- Department of Biochemistry, Microbiology, and Molecular Biology, the Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Science, the Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
35
|
Germline variability and tumor expression level of ribosomal protein gene RPL28 are associated with survival of metastatic colorectal cancer patients. Sci Rep 2019; 9:13008. [PMID: 31506518 PMCID: PMC6736932 DOI: 10.1038/s41598-019-49477-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
This study investigated the potential of single nucleotide polymorphisms as predictors of survival in two cohorts comprising 417 metastatic colorectal cancer (mCRC) patients treated with the FOLFIRI (folinic acid, 5-fluorouracil and irinotecan) regimen. The rs4806668G > T of the ribosomal protein gene RPL28 was associated with shorter progression-free survival and overall survival by 5 and 9 months (P = 0.002), with hazard ratios of 3.36 (P < 0.001) and 3.07 (P = 0.002), respectively. The rs4806668T allele was associated with an increased RPL28 expression in transverse normal colon tissues (n = 246, P = 0.007). RPL28 expression was higher in colorectal tumors compared to paired normal tissues by up to 124% (P < 0.001) in three independent datasets. Metastatic cases with highest RPL28 tumor expression had a reduced survival in two datasets (n = 88, P = 0.009 and n = 56, P = 0.009). High RPL28 was further associated with changes in immunoglobulin and extracellular matrix pathways. Repression of RPL28 reduced proliferation by 1.4-fold to 5.6-fold (P < 0.05) in colon cancer HCT116 and HT-29 cells. Our findings suggest that the ribosomal RPL28 protein may influence mCRC outcome.
Collapse
|
36
|
Zhang S, Chatterjee T, Godoy C, Wu L, Liu QJ, Carmon KS. GPR56 Drives Colorectal Tumor Growth and Promotes Drug Resistance through Upregulation of MDR1 Expression via a RhoA-Mediated Mechanism. Mol Cancer Res 2019; 17:2196-2207. [PMID: 31444231 DOI: 10.1158/1541-7786.mcr-19-0436] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/19/2019] [Accepted: 08/20/2019] [Indexed: 12/26/2022]
Abstract
Drug resistance continues to be a major obstacle of effective therapy for colorectal cancer, leading to tumor relapse or treatment failure. Cancer stem cells (CSC) or tumor-initiating cells are a subpopulation of tumor cells which retain the capacity for self-renewal and are suggested to be implicated in drug resistance. LGR5 is highly expressed in colorectal cancer and marks CSCs that drive tumor growth and metastasis. LGR5(+) CSCs cells were shown to interconvert with more drug-resistant LGR5(-) cancer cells, and treatment with LGR5-targeted antibody-drug conjugates (ADC) eliminated LGR5(+) tumors, yet a fraction of LGR5(-) tumors eventually recurred. Therefore, it is important to identify mechanisms associated with CSC plasticity and drug resistance in order to develop curative therapies. Here, we show that loss of LGR5 in colon cancer cells enhanced resistance to irinotecan and 5-fluorouracil and increased expression of adhesion G-protein-coupled receptor, GPR56. GPR56 expression was significantly higher in primary colon tumors versus matched normal tissues and correlated with poor survival outcome. GPR56 enhanced drug resistance through upregulation of MDR1 levels via a RhoA-mediated signaling mechanism. Loss of GPR56 led to suppression of tumor growth and increased sensitivity of cancer cells to chemotherapy and monomethyl auristatin E-linked anti-LGR5 ADCs, by reducing MDR1 levels. These findings suggest that upregulation of GPR56 may be a mechanism associated with CSC plasticity by which LGR5(-) cancer cells acquire a more drug-resistant phenotype. IMPLICATIONS: Our findings suggest that targeting GPR56 may provide a new strategy for the treatment of colorectal cancer and combatting drug resistance.
Collapse
Affiliation(s)
- Sheng Zhang
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Treena Chatterjee
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Carla Godoy
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ling Wu
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Qingyun J Liu
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Kendra S Carmon
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
37
|
Sun R, Basu S, Zeng M, Sunsong R, Li L, Ghose R, Wang W, Liu Z, Hu M, Gao S. Xiao-Chai-Hu-Tang (XCHT) Intervening Irinotecan’s Disposition: The Potential of XCHT in Alleviating Irinotecan-Induced Diarrhea. Curr Cancer Drug Targets 2019; 19:551-560. [DOI: 10.2174/1568009618666181029153255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/23/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022]
Abstract
<P>Background: Diarrhea is a severe side effect of irinotecan, a pro-drug of SN-38 used for the treatment of many types of cancers. Pre-clinical and clinical studies showed that decreasing the colonic exposure of SN-38 can mitigate irinotecan-induced diarrhea. </P><P> Objective: The purpose of this study is to evaluate the anti-diarrhea potential of Xiao-Chai-Hu-Tang (XCHT), a traditional Chinese herbal formula, against irinotecan-induced diarrhea by determining if and how XCHT alters the disposition of SN-38. </P><P> Methods: LC-MS/MS was used to quantify the concentrations of irinotecan and its major metabolites (i.e., SN-38, SN-38G). An Intestinal perfusion model was used to determine the effect of XCHT on the biliary and intestinal secretions of irinotecan, SN-38, and SN-38G. Pharmacokinetic (PK) studies were performed to determine the impact of XCHT on the blood and fecal concentrations of irinotecan, SN-38, and SN-38G. </P><P> Results: The results showed that XCHT significantly inhibits both biliary and intestinal excretions of irinotecan, SN-38, and SN-38G (range: 35% to 95%). PK studies revealed that the fecal concentrations of irinotecan and SN-38 were significantly decreased from 818.35 ± 120.2 to 411.74 ± 138.83 µg/g or from 423.95 ± 76.44 to 245.63 ± 56.72 µg/g (p<0.05) by XCHT, respectively, suggesting the colonic exposure of SN-38 is significantly decreased by XCHT. PK studies also showed that the plasma concentrations of irinotecan, SN-38, and SN-38G were not affected by XCHT. </P><P> Conclusion: In conclusion, XCHT significantly decreased the exposure of SN-38 in the gut without affecting its plasma level, thereby possessing the potential of alleviating irinotecan-induced diarrhea without negatively impacting its therapeutic efficacy.</P>
Collapse
Affiliation(s)
- Rongjin Sun
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 51006, China
| | - Sumit Basu
- Department of Pharmacological and Pharmaceutical Sciences, The University of Houston, 1441 Moursund Street, Houston, TX 77030, United States
| | - Min Zeng
- Department of Pharmacological and Pharmaceutical Sciences, The University of Houston, 1441 Moursund Street, Houston, TX 77030, United States
| | - Robin Sunsong
- Department of Pharmaceutical and Environmental Sciences, Texas Southern University, 3100 Cleburne Street, Houston, TX 77004, United States
| | - Li Li
- Department of Pharmacological and Pharmaceutical Sciences, The University of Houston, 1441 Moursund Street, Houston, TX 77030, United States
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, The University of Houston, 1441 Moursund Street, Houston, TX 77030, United States
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, The University of Houston, 1441 Moursund Street, Houston, TX 77030, United States
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 51006, China
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, The University of Houston, 1441 Moursund Street, Houston, TX 77030, United States
| | - Song Gao
- Department of Pharmaceutical and Environmental Sciences, Texas Southern University, 3100 Cleburne Street, Houston, TX 77004, United States
| |
Collapse
|
38
|
Xing J, Zhang X, Wang Z, Zhang H, Chen P, Zhou G, Sun C, Gu N, Ji M. Novel lipophilic SN38 prodrug forming stable liposomes for colorectal carcinoma therapy. Int J Nanomedicine 2019; 14:5201-5213. [PMID: 31371956 PMCID: PMC6634269 DOI: 10.2147/ijn.s204965] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/27/2019] [Indexed: 01/26/2023] Open
Abstract
Background: SN38 (7-ethyl-10-hydroxy camptothecin), as a potent metabolite of irinotecan, is highly efficacious in cancer treatment. However, the clinical utility of SN38 has been greatly limited due to its undesirable properties, such as poor solubility and low stability. Materials and methods: In order to overcome these weaknesses, moeixitecan, a lipophilic SN38 prodrug containing a SN-38, a trolox, a succinic acid linker, and a hexadecanol chain, was loaded into liposomal nanoparticles by ethanol injection method. Results: Experiments showed that the moeixitecan-loaded liposomal nanoparticles (MLP) with a diameter of 105.10±1.49 nm have a satisfactory drug loading rate (90.54±0.41%), high solubility and stability, and showed sustained release of SN38. Notably, MLP exhibited better antitumor activity against human colon adenocarcinoma cells than irinotecan, a FDA-approved drug for the treatment of advanced colorectal cancer. Furthermore, xenograft model results showed that MLP outperformed irinotecan in terms of pharmacokinetics, in vivo therapeutic efficacy and safety. Finally, we used molecular dynamic simulations to explore the association between the structure of MLP and the physical and functional properties of MLP, moeixitecan molecules in MLP folded themselves inside the hydrocarbon chain of the lipid bilayer, which led an increased acyl chain order of the lipid bilayer, and therefore enhanced the lactone ring stability protecting it from hydrolysis. Conclusion: Our MLP constructing strategy by liposome engineering technology may serve a promising universal approach for the effective and safe delivery of lipophilic prodrug.
Collapse
Affiliation(s)
- Jing Xing
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China.,School of Biological Science and Medical Engineering and Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Suzhou 215123, People's Republic of China
| | - Xiquan Zhang
- Nanjing Institute of Pharmaceutical Research and Development, Chia-Tai Tianqing Pharmaceutical Group Co. Ltd, Nanjing 210023, People's Republic of China
| | - Zhe Wang
- Emergency Department, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Huanqing Zhang
- Nanjing Institute of Pharmaceutical Research and Development, Chia-Tai Tianqing Pharmaceutical Group Co. Ltd, Nanjing 210023, People's Republic of China
| | - Peng Chen
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China.,School of Biological Science and Medical Engineering and Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Suzhou 215123, People's Republic of China
| | - Gaoxin Zhou
- School of Biomedical Engineering, Shenzhen University, Shenzhen 518071, People's Republic of China
| | - Chunlong Sun
- College of Biological and Environmental Engineering, Binzhou University, Binzhou 256603, People's Republic of China
| | - Ning Gu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China.,School of Biological Science and Medical Engineering and Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Suzhou 215123, People's Republic of China
| | - Min Ji
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, People's Republic of China.,School of Biological Science and Medical Engineering and Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Suzhou 215123, People's Republic of China
| |
Collapse
|
39
|
Sugarman R, Patel R, Sharma S, Plenker D, Tuveson D, Saif MW. Pharmacokinetics and pharmacodynamics of new drugs for pancreatic cancer. Expert Opin Drug Metab Toxicol 2019; 15:541-552. [PMID: 31241371 DOI: 10.1080/17425255.2019.1637417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: Pancreatic cancer (PC) remains a disease with a dismal prognosis. Despite accounting for only 3% of cancer diagnosis, 7% of all cancer deaths in the United States are from PC. This is explained by many being diagnosed with late-stage disease and the cancer's resistance to chemotherapy. Since 1996 there have only been two upfront regimens found to be superior to gemcitabine, FOLFIRINOX (5-fluorouracil/leucovorin and oxaliplatin) and gemcitabine plus nab-paclitaxel. Areas covered: Clinical pharmacology of newer agents that are either approved or being investigated in the management of PC. Knowledge of their pharmacokinetics, pharmacodynamics, and pharmacogenetics can be used to predict outcomes for specific patient populations. Drugs discussed include nanoliposomal irinotecan, pegvorhyaluronidase alfa, poly (ADP-ribose) polymerase enzyme inhibitors, larotrectinib, and napabucasin. Expert opinion: PC is a heterogeneous disease and outcomes are likely to improve as better predictive models of an individual's response to different therapies are developed. This may be best accomplished through phase 0 studies and the use of tumor organoid models grown from initial biopsies or resected tissue. The genetic and physical makeup of the tumor as well as the functional characterization in patient-derived organoids (PDOs), can help guide which agents may be most efficacious or toxic.
Collapse
Affiliation(s)
- Ryan Sugarman
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| | - Rajvi Patel
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| | - Sandhya Sharma
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| | - Dennis Plenker
- b Cold Spring Harbor Laboratory , Cold Spring Harbor , NY , USA
| | - David Tuveson
- b Cold Spring Harbor Laboratory , Cold Spring Harbor , NY , USA
| | - Muhammad Wasif Saif
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| |
Collapse
|
40
|
Chamseddine AN, Ducreux M, Armand JP, Paoletti X, Satar T, Paci A, Mir O. Intestinal bacterial β-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Pharmacol Ther 2019; 199:1-15. [DOI: 10.1016/j.pharmthera.2019.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
41
|
Abstract
The microbiome is emerging as a prominent factor affecting human health, and its dysbiosis is associated with various diseases. Compositional profiling of microbiome is increasingly being supplemented with functional characterization. Metaproteomics is intrinsically focused on functional changes and therefore will be an important tool in those studies of the human microbiome. In the past decade, development of new experimental and bioinformatic approaches for metaproteomics has enabled large-scale human metaproteomic studies. However, challenges still exist, and there remains a lack of standardizations and guidelines for properly performing metaproteomic studies on human microbiome. Herein, we provide a perspective of recent developments, the challenges faced, and the future directions of metaproteomics and its applications. In addition, we propose a set of guidelines/recommendations for performing and reporting the results from metaproteomic experiments for the study of human microbiomes. We anticipate that these guidelines will be optimized further as more metaproteomic questions are raised and addressed, and metaproteomic applications are published, so that they are eventually recognized and applied in the field.
Collapse
Affiliation(s)
- Xu Zhang
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| | - Daniel Figeys
- Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine , University of Ottawa , Ottawa , Ontario K1H 8M5 , Canada
| |
Collapse
|
42
|
Tang L, Li X, Wan L, Xiao Y, Zeng X, Ding H. Herbal Medicines for Irinotecan-Induced Diarrhea. Front Pharmacol 2019; 10:182. [PMID: 30983992 PMCID: PMC6450188 DOI: 10.3389/fphar.2019.00182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/13/2019] [Indexed: 12/12/2022] Open
Abstract
Irinotecan (CPT-11), a water-soluble derivative of camptothecin, belongs to the class of DNA topoisomerase I inhibitors and has been approved worldwide for the treatment of advanced colorectal cancer, lung cancer, and malignant lymphoma. Although CPT-11-based chemotherapy is widely used, severe gastrointestinal (GI) toxicity, especially late-onset diarrhea, is a common adverse reaction, limiting clinical application of the drug. The incidence of grade 3 or 4 diarrhea is high, with 20-40% of CPT-11-treated patients experiencing this adverse effect. High-dose loperamide and octreotide are generally recommended for treatment of CPT-11-induced diarrhea. However, in clinical practice, loperamide is associated with a significant failure rate and the beneficial effects of octreotide are controversial. An accumulating number of recent studies have suggested that medicinal herbs and their derived phytocompounds may be effective complementary treatments for CPT-11-induced diarrhea. In this mini-review, we briefly summarize currently available literatures regarding the formulae and herbs/natural products used as adjuvants in animal and clinical studies for the treatment of diarrhea caused by CPT-11.
Collapse
Affiliation(s)
- Liu Tang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xiaolei Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Liping Wan
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Yao Xiao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xin Zeng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
43
|
Yijie Q, Xuejun W, Yuanyao X, Yongqiong N, Jianxin Y, Yabin D, Xiangyang L. Effect of X-ray irradiation on pharmacokinetics of irinotecan hydrochloride and expression of CES1 and CYP3A1 in rats. Fundam Clin Pharmacol 2019; 33:558-566. [PMID: 30811654 DOI: 10.1111/fcp.12456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/05/2019] [Accepted: 02/25/2019] [Indexed: 12/30/2022]
Abstract
Concurrent chemoradiation with irinotecan hydrochloride (CPT-11) is accepted for cancer treatment. However, the effects of X-ray irradiation on chemotherapeutics in the plasma remain unclear. We evaluated the pharmacokinetics of CPT-11 in rats after exposure to X-ray irradiation and examined the changes of protein and mRNA expression of CES1 and CYP3A1. The X-ray irradiation with 1 Gy and 5 Gy was delivered to the whole body of rats. CPT-11 at 30 and 60 mg/kg, respectively, was intravenously infused 24 h after irradiation. CPT-11 was determined by RP-HPLC in plasma. ELISA and PCR were used to analyze the protein and mRNA expression of CES1 and CYP3A1, respectively. Compared with control rats, the X-ray irradiation decreased the AUC of CPT-11 (30 mg/kg) by 15.6% at 1 Gy and 39.0% at 5 Gy and increased the CL by 60.0% at 5 Gy. The X-ray irradiation could also decrease the AUC of CPT-11 (60 mg/kg) and increase the CL. In addition, the protein and mRNA expression of CES1 and CYP3A1 were increased significantly in rats after irradiation. This study found significant changes in the pharmacokinetics of CPT-11 in rats after exposure to X-ray irradiation, and they might be due to significant increases in the expressions of CYP3A1 and CES1. The pharmacokinetics of CPT-11 should be rechecked, and the optimal CPT-11 dose should be reevaluated during concurrent chemoradiation therapy.
Collapse
Affiliation(s)
- Qiao Yijie
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, 810016, China
| | - Wang Xuejun
- Department of Radiology, Red Cross Hospital of Qinghai, Xining, 810001, China
| | - Xin Yuanyao
- College of Eco-Environmental Engineering, Qinghai University, Xining, 810016, China
| | - Nian Yongqiong
- College of Eco-Environmental Engineering, Qinghai University, Xining, 810016, China
| | - Yang Jianxin
- Medical College, Qinghai University, Xining, 810001, China
| | - Duan Yabin
- Medical College, Qinghai University, Xining, 810001, China
| | - Li Xiangyang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, 810016, China
| |
Collapse
|
44
|
Cho PJ, Kim JH, Lee HS, Kim JA, Lee S. Identification of specific UGT1A9-mediated glucuronidation of licoricidin in human liver microsomes. Biopharm Drug Dispos 2019; 40:94-98. [DOI: 10.1002/bdd.2169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/29/2018] [Accepted: 12/16/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Pil Joung Cho
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu 41566 Republic of Korea
| | - Ju-Hyun Kim
- College of Pharmacy; Yeungnam University; Gyeongsan 38541 Republic of Korea
| | - Hye Suk Lee
- BK21 Plus Team for Creative Leader Program for Pharmacomics-based Future, Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy; The Catholic University of Korea; Bucheon 14662 Republic of Korea
| | - Jeong Ah Kim
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu 41566 Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences; Kyungpook National University; Daegu 41566 Republic of Korea
| |
Collapse
|
45
|
Chen F, Cai Y, Huang L, Chen Y, Luo X. Synthesis of a SN38 prodrug grafted to amphiphilic phosphorylcholine polymers and their prodrug miceller properties. NEW J CHEM 2019. [DOI: 10.1039/c8nj04908d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polymer prodrug micelles, combining the advantages of prodrugs and polymer micelles, can greatly improve the solubility, permeability and stability of drugs.
Collapse
Affiliation(s)
- Fan Chen
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Yuanyuan Cai
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Lei Huang
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Yuanwei Chen
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Xianglin Luo
- College of Polymer Science and Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
- State Key Lab of Polymer Materials Engineering
| |
Collapse
|
46
|
Liu Y, Li Y, Li J, Xie Z, Wang Y, Chen Z. Ethyl violet-bovine serum albumin fluorescent protein nanovessels target to lysosomes and mitochondria. Nanomedicine (Lond) 2018; 14:19-31. [PMID: 30547703 DOI: 10.2217/nnm-2018-0281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Organelles are essential in maintaining homeostasis of mammalian cells. Monitoring the morphology and dynamics of organelles is of significance in cell state determination and disease diagnosis. MATERIALS & METHODS We describe here a new material called ethyl violet-bovine serum albumin fluorescent protein nanovessel (EV-BSA FPN). Upon heating, BSA was denatured to form higher polyhedral structures, which was prone to EV binding. These dye-protein hybrid materials were red fluorescence emissive upon excitation. RESULTS EV-BSA FPNs can be readily internalized by mammalian cells and dual localized in lysosomes and mitochondria. Besides, EV-BSA FPN can serve as carriers and efficiently deliver drug into cells. CONCLUSION EV-BSA FPNs can be dual function fluorescent vessels for both dual-organelle imaging and drug delivery.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Supramolecular Structure & Materials & Institute of Theoretical Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, PR China
| | - Yuanyuan Li
- State Key Laboratory of Polymer Physics & Chemistry, The First Hospital of Jilin University, Xinmin Street, Changchun, Jilin 130021, PR China
| | - Jia Li
- Beijing Key Laboratory of Gene Resource & Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics & Chemistry, The First Hospital of Jilin University, Xinmin Street, Changchun, Jilin 130021, PR China
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource & Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, PR China
| | - Zhijun Chen
- State Key Laboratory of Supramolecular Structure & Materials & Institute of Theoretical Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, PR China
| |
Collapse
|
47
|
Characterization of new, efficient Mycobacterium tuberculosis topoisomerase-I inhibitors and their interaction with human ABC multidrug transporters. PLoS One 2018; 13:e0202749. [PMID: 30183750 PMCID: PMC6124754 DOI: 10.1371/journal.pone.0202749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/08/2018] [Indexed: 12/22/2022] Open
Abstract
Drug resistant tuberculosis (TB) is a major worldwide health problem. In addition to the bacterial mechanisms, human drug transporters limiting the cellular accumulation and the pharmacological disposition of drugs also influence the efficacy of treatment. Mycobacterium tuberculosis topoisomerase-I (MtTopo-I) is a promising target for antimicrobial treatment. In our previous work we have identified several hit compounds targeting the MtTopo-I by in silico docking. Here we expand the scope of the compounds around three scaffolds associated with potent MtTopo-I inhibition. In addition to measuring the effect of newly generated compounds on MtTopo-I activity, we characterized the compounds’ antimicrobial activity, toxicity in human cells, and interactions with human multidrug transporters. Some of the newly developed MtTopo-I inhibitors have strong antimicrobial activity and do not harm mammalian cells. Moreover, our studies revealed significant human ABC drug transporter interactions for several MtTopo-I compounds that may modify their ADME-Tox parameters and cellular effects. Promising new drug candidates may be selected based on these studies for further anti-TB drug development.
Collapse
|
48
|
Lucić Vrdoljak A, Fuchs N, Mikolić A, Žunec S, Brčić Karačonji I, Jurič A, Prester L, Micek V, Neuberg M, Čanović S, Mršić G, Kopjar N. Irinotecan and Δ⁸-Tetrahydrocannabinol Interactions in Rat Liver: A Preliminary Evaluation Using Biochemical and Genotoxicity Markers. Molecules 2018; 23:E1332. [PMID: 29865166 PMCID: PMC6100385 DOI: 10.3390/molecules23061332] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/23/2018] [Accepted: 05/31/2018] [Indexed: 01/27/2023] Open
Abstract
There is growing interest regarding the use of herbal preparations based on Cannabis sativa for medicinal purposes, despite the poorly understood interactions of their main constituent Δ⁸-tetrahydrocannabinol (THC) with conventional drugs, especially cytostatics. The objective of this pilot study was to prove whether the concomitant intake of THC impaired liver function in male Wistar rats treated with the anticancer drug irinotecan (IRI), and evaluate the toxic effects associated with this exposure. IRI was administered once intraperitoneally (at 100 mg/kg of the body weight (b.w.)), while THC was administered per os repeatedly for 1, 3, and 7 days (at 7 mg/kg b.w.). Functional liver impairments were studied using biochemical markers of liver function (aspartate aminotransferase-AST, alanine aminotransferase-ALP, alkaline phosphatase-AP, and bilirubin) in rats given a combined treatment, single IRI, single THC, and control groups. Using common oxidative stress biomarkers, along with measurement of primary DNA damage in hepatocytes, the degree of impairments caused at the cellular level was also evaluated. THC caused a time-dependent enhancement of acute toxicity in IRI-treated rats, which was confirmed by body and liver weight reduction. Although single THC affected ALP and AP levels more than single IRI, the levels of liver function markers measured after the administration of a combined treatment mostly did not significantly differ from control. Combined exposure led to increased oxidative stress responses in 3- and 7-day treatments, compared to single IRI. Single IRI caused the highest DNA damage at all timepoints. Continuous 7-day oral exposure to single THC caused an increased mean value of comet tail length compared to its shorter treatments. Concomitant intake of THC slightly affected the levels of IRI genotoxicity at all timepoints, but not in a consistent manner. Further studies are needed to prove our preliminary observations, clarify the underlying mechanisms behind IRI and THC interactions, and unambiguously confirm or reject the assumptions made herein.
Collapse
Affiliation(s)
- Ana Lucić Vrdoljak
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| | - Nino Fuchs
- University Hospital Centre Zagreb, HR-10000 Zagreb, Croatia.
| | - Anja Mikolić
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| | - Suzana Žunec
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| | | | - Andreja Jurič
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| | - Ljerka Prester
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| | - Vedran Micek
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| | - Marijana Neuberg
- University Centre Varaždin, University North, HR-42000 Varaždin, Croatia.
| | | | - Gordan Mršić
- Forensic Science Centre "Ivan Vučetić", HR-10000 Zagreb, Croatia.
| | - Nevenka Kopjar
- Institute for Medical Research and Occupational Health, HR-10001 Zagreb, Croatia.
| |
Collapse
|
49
|
Palmirotta R, Carella C, Silvestris E, Cives M, Stucci SL, Tucci M, Lovero D, Silvestris F. SNPs in predicting clinical efficacy and toxicity of chemotherapy: walking through the quicksand. Oncotarget 2018; 9:25355-25382. [PMID: 29861877 PMCID: PMC5982750 DOI: 10.18632/oncotarget.25256] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/07/2018] [Indexed: 12/19/2022] Open
Abstract
In the "precision medicine" era, chemotherapy still remains the backbone for the treatment of many cancers, but no affordable predictors of response to the chemodrugs are available in clinical practice. Single nucleotide polymorphisms (SNPs) are gene sequence variations occurring in more than 1% of the full population, and account for approximately 80% of inter-individual genomic heterogeneity. A number of studies have investigated the predictive role of SNPs of genes enrolled in both pharmacodynamics and pharmacokinetics of chemotherapeutics, but the clinical implementation of related results has been modest so far. Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. While formal recommendations for routine testing of these SNPs cannot be drawn at this moment, therapeutic decisions may indeed benefit of germline genomic information, when available. Here, we summarize the clinical impact of germline genomic variants on the efficacy and toxicity of major chemodrugs, with the aim to facilitate the therapeutic expectance of clinicians in the odiern quicksand field of complex molecular biology concepts and controversial trial data interpretation.
Collapse
Affiliation(s)
- Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Claudia Carella
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Erica Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Mauro Cives
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Stefania Luigia Stucci
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Marco Tucci
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Domenica Lovero
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
50
|
Wang L, Chan CEL, Wong ALA, Wong FC, Lim SW, Chinnathambi A, Alharbi SA, Lee LSU, Soo R, Yong WP, Lee SC, Ho PCL, Sethi G, Goh BC. Combined use of irinotecan with histone deacetylase inhibitor belinostat could cause severe toxicity by inhibiting SN-38 glucuronidation via UGT1A1. Oncotarget 2018; 8:41572-41581. [PMID: 28157715 PMCID: PMC5522258 DOI: 10.18632/oncotarget.15017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/07/2017] [Indexed: 12/27/2022] Open
Abstract
SN-38, the active metabolite of irinotecan, and histone deacetylase inhibitors (HDACis) such as belinostat, vorinostat and panobinostat, have all been shown to be deactivated by glucuronidation via UGTs. Since they all compete for UGTs for deactivation, we aimed to investigate the inhibitory effect of various HDACis on the glucuronidation of SN-38. This inhibitory effect was determined by measuring the formation rate of SN-38 glucuronide after SN-38 incubation with human recombinant UGT1A isoforms (1A1, 1A6, 1A7 and 1A9) and pooled human liver microsomes (HLM, wild type, UGT1A1*1*28 and UGT1A1*28*28 allelic variants), with and without HDACis. The data showed that belinostat at 100 and 200 µmol/L inhibited SN-38 glucuronidation via UGT1A1 in a dose-dependent manner, causing significant decrease in Vmax and CLint (p < 0.05) from 12.60 to 1.95 pmol/min/mg and 21.59 to 4.20 μL/min/mg protein respectively. Similarly, in HLMs, Vmax dropped from 41.13 to 10.54, 24.96 to 3.77 and 6.23 to 3.30 pmol/min/mg, and CLint reduced from 81.25 to 26.11, 29.22 to 6.10 and 5.40 to 1.34 µL/min/mg protein for the respective wild type, heterozygous and homozygous variants. Interestingly, belinostat at 200 µmol/L that is roughly equivalent to the average Cmax, 183 µmol/L of belinostat at a dose of 1,400 mg/m2 given intravenously once per day on days 1 to 5 every 3 weeks, was able to inhibit both heterozygous and homozygous variants to same extents (~64%). This highlights the potential clinical significance, as a large proportion of patients could be at risk of developing severe toxicity if irinotecan is co-administered with belinostat.
Collapse
Affiliation(s)
- Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, National University Health System, Singapore
| | - Chong En Linus Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacy, National University of Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Fang Cheng Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Siew Woon Lim
- Department of Pharmacy, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | | | - Ross Soo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Soo Chin Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, National University Health System, Singapore.,Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth WA, Australia
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, National University Health System, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| |
Collapse
|