1
|
Razavi SZ, Amini-Khoei H, Rahimi-Madiseh M, Bijad E, Lorigooini Z. Modulation of neuroinflammation and oxidative stress by Echinacea purpurea extract: Therapeutic potential in maternal separation-induced autism spectrum disorder. J Psychiatr Res 2025; 184:118-127. [PMID: 40049118 DOI: 10.1016/j.jpsychires.2025.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/21/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is defined by ongoing problems in social interaction and communication and repetitive, constrained behavior patterns. The link between oxidative stress (OS) and inflammation with ASD has been shown in previous studies. E. purpurea is well-known for its potential antioxidant and anti-inflammatory pharmacological properties. In this study, we aimed to evaluate the effects of E. purpurea hydroalcoholic extract on autistic-like behaviors following a mouse model of maternal separation (MS) stress, focusing on possible anti-neuroinflammation and antioxidative stress. METHODS 70% hydro-ethanolic extract was macerated from the aerial parts of E. purpurea. Standardization was done by determining the amount of chicoric acid in the extract using the UHPLC method. Then, behavioral analysis was done on 75 male mice that underwent MS. Mice were treated with normal saline or 75, 150, and 300 mg/kg of the extract. Sociability behaviors and stereotyping behaviors have been evaluated. Also, their total antioxidant capacity (TAC), nitrite levels, and malondialdehyde (MDA) were measured in the hippocampus. In addition, the expression of inflammatory factors, including interleukin-1 (IL-1), NLRP3, and TLR4, has been determined by quantitative real-time PCR (qRT-PCR). Data were analyzed after collection using PRISM statistical software. RESULTS Our findings indicated that MS caused autistic-like behaviors in mice (increased sociability index and social preference index) and increased repetitive behaviors (increased number of buried marbles). These autistic-like behaviors are associated with increased MDA, nitrite, over-expression of inflammatory genes, decreased MDA, nitrite, over-expression of inflammatory genes, and decreased TAC in the hippocampus. E. purpurea extract significantly reversed these adverse effects of MS. CONCLUSION The results of this study showed that E. purpurea extract might reduce autistic-like behaviors in MS by attenuating neuroinflammation and oxidative stress states.
Collapse
Affiliation(s)
- Seyedeh Zahra Razavi
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Rahimi-Madiseh
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Bijad
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
2
|
Yamaguchi Y, Takeda K, Yoshida S, Maruo K. Optimal biological dose selection in dose-finding trials with model-assisted designs based on efficacy and toxicity: a simulation study. J Biopharm Stat 2024; 34:379-393. [PMID: 37114985 DOI: 10.1080/10543406.2023.2202259] [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: 11/18/2022] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
With the emergence of molecular targeted agents and immunotherapies in anti-cancer treatment, a concept of optimal biological dose (OBD), accounting for efficacy and toxicity in the framework of dose-finding, has been widely introduced into phase I oncology clinical trials. Various model-assisted designs with dose-escalation rules based jointly on toxicity and efficacy are now available to establish the OBD, where the OBD is generally selected at the end of the trial using all toxicity and efficacy data obtained from the entire cohort. Several measures to select the OBD and multiple methods to estimate the efficacy probability have been developed for the OBD selection, leading to many options in practice; however, their comparative performance is still uncertain, and practitioners need to take special care of which approaches would be the best for their applications. Therefore, we conducted a comprehensive simulation study to demonstrate the operating characteristics of the OBD selection approaches. The simulation study revealed key features of utility functions measuring the toxicity-efficacy trade-off and suggested that the measure used to select the OBD could vary depending on the choice of the dose-escalation procedure. Modelling the efficacy probability might lead to limited gains in OBD selection.
Collapse
Affiliation(s)
- Yusuke Yamaguchi
- Astellas Pharma Global Development, Inc, Northbrook, Illinois, USA
| | - Kentaro Takeda
- Astellas Pharma Global Development, Inc, Northbrook, Illinois, USA
| | | | - Kazushi Maruo
- Department of Biostatistics, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
3
|
Rahat MM, Sabtan H, Simanovich E, Haddad A, Gazitt T, Feld J, Slobodin G, Kibari A, Elias M, Zisman D, Rahat MA. Soluble CD147 regulates endostatin via its effects on the activities of MMP-9 and secreted proteasome 20S. Front Immunol 2024; 15:1319939. [PMID: 38318187 PMCID: PMC10840997 DOI: 10.3389/fimmu.2024.1319939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
During progression of rheumatoid arthritis (RA), angiogenesis provides oxygen and nutrients for the cells' increased metabolic demands and number. To turn on angiogenesis, pro-angiogenic factors must outweigh anti-angiogenic factors. We have previously shown that CD147/extracellular matrix metalloproteinase inducer (EMMPRIN) can induce the expression of the pro-angiogenic factors vascular endothelial growth factor (VEGF) and matrix metallopeptidase 9 (MMP-9) in a co-culture of the human HT1080 fibrosarcoma and U937 monocytic-like cell lines. However, whether CD147 influences anti-angiogenic factors was not known. We now show that relative to single cultures, the co-culture of these cells not only enhanced pro-angiogenic factors but also decreased the anti-angiogenic factors endostatin and thrombospondin-1 (Tsp-1), generally increasing the angiogenic potential as measured by a wound assay. Using anti-CD147 antibody, CD147 small interfering RNA (siRNA), and recombinant CD147, we demonstrate that CD147 hormetically regulates the generation of endostatin but has no effect on Tsp-1. Since endostatin is cleaved from collagen XVIII (Col18A), we applied different protease inhibitors and established that MMP-9 and proteasome 20S, but not cathepsins, are responsible for endostatin generation. MMP-9 and proteasome 20S collaborate to synergistically enhance endostatin generation, and in a non-cellular system, CD147 enhanced MMP-9 activity and hormetically regulated proteasome 20S activity. Serum samples obtained from RA patients and healthy controls mostly corroborated these findings, indicating clinical relevance. Cumulatively, these findings suggest that secreted CD147 mediates a possibly allosteric effect on MMP-9 and proteasome 20S activities and can serve as a switch that turns angiogenesis on or off, depending on its ambient concentrations in the microenvironment.
Collapse
Affiliation(s)
- Maya M. Rahat
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel
| | - Hala Sabtan
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
| | | | - Amir Haddad
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tal Gazitt
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Joy Feld
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Gleb Slobodin
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Rheumatology, Bnai Zion Medical Center, Haifa, Israel
| | - Adi Kibari
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Muna Elias
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
| | - Devy Zisman
- Department of Rheumatology, Carmel Medical Center, Haifa, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Michal A. Rahat
- Immunotherapy Laboratory, Carmel Medical Center, Haifa, Israel
- The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
4
|
Wu S, Zhang Q, Li Y, Liang H. Assessment of nonlinear dose-response relationships via nonparametric regression. J Biopharm Stat 2024; 34:136-145. [PMID: 36861953 DOI: 10.1080/10543406.2023.2183505] [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: 07/18/2022] [Accepted: 01/20/2023] [Indexed: 03/03/2023]
Abstract
We propose a simple approach to assess whether a nonlinear parametric model is appropriate to depict the dose-response relationships and whether two parametric models can be applied to fit a dataset via nonparametric regression. The proposed approach can compensate for the ANOVA, which is sometimes conservative, and is very easy to implement. We illustrate the performance by analyzing experimental examples and a small simulation study.
Collapse
Affiliation(s)
- Shunyao Wu
- Department of Computer Science and Technology, Qingdao University, Shandong, Qingdao, China
| | - Qi Zhang
- Department of Computer Science and Technology, Qingdao University, Shandong, Qingdao, China
| | - Yuanzhang Li
- Department of Statistics, George Washington University, DC, Washington, USA
| | - Hua Liang
- Department of Statistics, George Washington University, DC, Washington, USA
| |
Collapse
|
5
|
Walson PD. Personalized Minimal Effective Concentration Therapy. Clin Ther 2023; 45:1289-1292. [PMID: 37838561 DOI: 10.1016/j.clinthera.2023.09.015] [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: 05/08/2023] [Revised: 07/22/2023] [Accepted: 09/17/2023] [Indexed: 10/16/2023]
Abstract
It has been recognized for literally centuries that patients should be given only the amount of medication necessary to treat disease(s) or relieve symptoms. It is also well known that this amount can vary greatly between patients or even over time in the same patient. The ability to identify this amount, that is, to "personalize" dosing, requires a reliable measure of a patient's response to treatment. The development of analytical methods for the accurate measurement of pharmacologically meaningful drug concentrations in physiologic fluids, combined with mathematical methods for reliable prediction of how dosing changes affect these concentrations, has led to the development of therapeutic drug management (TDM) for more effective individualization of dosing. Using TDM, clinicians modify dosing to achieve concentrations or exposures (ie, AUC) found to be effective in patients with similar clinical attributes and conditions. These concentrations, called therapeutic (or target) concentrations or exposure ranges (TRs), are specific to both disease/condition and patient population. TDM is routinely used by many clinicians to adjust dosing of a wide range of medications for maximal efficacy and limited toxicity, thereby improving clinical outcomes. Failure to properly perform TDM or to appreciate the limitations of TDM have, however, contributed to the delayed acceptance of TDM by clinicians. This Commentary briefly discusses the limitations and the benefits of TR-guided TDM, and then discusses immunosuppressant drugs and anticancer medications as examples of drugs that require clinicians to change their prescribing practices from giving all patients the same or maximal tolerated doses, to instead adjusting individual doses to achieve minimal effective concentrations identified using circulating tumor- or graft-derived DNA or copy number instability rather than published TRs.
Collapse
Affiliation(s)
- Philip D Walson
- Department of Clinical Pharmacology, University Medical School Goettingen, Hannover, Germany.
| |
Collapse
|
6
|
Oliveira NMT, Dos Santos AE, Corso CR, Galindo CM, Adami ER, da Silva LCM, de Lima LTF, de Santana Filho AP, Dittrich RL, Klassen G, de Souza Ramos EA, Sassaki GL, Acco A. Chemical characterization and antineoplastic effect of oligosaccharides from Cabernet Franc red wine in mammary tumor model in mice. J Nutr Biochem 2023; 113:109253. [PMID: 36565967 DOI: 10.1016/j.jnutbio.2022.109253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
The present study characterized oligosaccharide compounds (Oligo) in Cabernet Franc red wine and investigated its antineoplastic effects against mammary tumor cells in vivo and in vitro, isolated or in combination with chemotherapy. The Oligo fraction was characterized by nuclear magnetic resonance spectroscopy and mass spectrometry. The complex mixture of Oligo showed high amounts of oligoxyloglucuronans, oligorhamnogalacturonans, oligoarabinogalactans, and oligoglucans, such as trehalose and isomaltotriose. To investigate the antineoplastic effects of Oligo, Female Swiss mice were subcutaneously inoculated with Ehrlich tumor cells and then received vehicle (distilled water, p.o.), Oligo solution (9, 35, or 70 mg/kg, p.o.), or methotrexate (1.5 mg/kg, i.p.). The treatments were administered in a conventional (21-d) or chemopreventive (42-d) protocol. Oligo reduced the growth of Ehrlich tumors in both protocols and increased the effectiveness of methotrexate in controlling tumor growth. Oligo did not reduce the viability of MCF-7, MDA-MB-231, MDA-MB-436, and HB4a human breast cells that were cultured for 48 h, showing no cytotoxicity. Overall, Oligo exerted an in vivo antineoplastic effect and modulated immune blood cells, dependent on treatment time, and was not directly cytotoxic to tumor cells. Thus, Oligo may indirectly regulate tumor cell development and may be a promising drug for cancer therapy in combination with methotrexate.
Collapse
Affiliation(s)
| | - André Eduardo Dos Santos
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Claudia Rita Corso
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | | | | | | | | | | | | | - Giseli Klassen
- Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Guilherme Lanzi Sassaki
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil.
| | - Alexandra Acco
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil.
| |
Collapse
|
7
|
Sharma S, Rana AK, Sharma A, Singh D. Inhibition of Mammalian Target of Rapamycin Attenuates Recurrent Seizures Associated Cardiac Damage in a Zebrafish Kindling Model of Chronic Epilepsy. J Neuroimmune Pharmacol 2022; 17:334-349. [PMID: 34537895 DOI: 10.1007/s11481-021-10021-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022]
Abstract
Sudden Unexpected Death in Epilepsy (SUDEP) is primarily linked with the cardiac irregularities that occur due to recurrent seizures. Our previous studies found a role of mTOR pathway activation in seizures-linked cardiac damage in a rat model. In continuation to the earlier work, the present study was devised to explore the role of rapamycin (mTOR inhibitor and clinically used immunosuppressive agent) in a zebrafish kindling model and associated cardiac damage. Adult zebrafish were incubated with increasing concentrations of rapamycin (1, 2 and, 4 μM), followed by pentylenetetrazole (PTZ) exposure to record seizure latency and severity. In another experiment, zebrafish were subjected to a standardized PTZ kindling protocol. The kindled fish were treated daily with rapamycin for up to 25 days, along with PTZ to record seizure severity. At the end, zebrafish heart was excised for carbonylation assay, gene expression, and protein quantification studies. In the acute PTZ convulsion test, treatment with rapamycin showed a significant increase in seizure latency and decreased seizure severity without any change in seizure incidence. Treatment with rapamycin also reduced the severity of seizures in kindled fish. The cardiac expressions of gpx, nppb, kcnh2, scn5a, mapk8, stat3, rps6 and ddit were decreased, whereas the levels of trxr2 and beclin 1 were increased following rapamycin treatment in kindled fish. Furthermore, rapamycin treatment also decreased p-mTOR expression and protein carbonyls level in the fish cardiac tissue. The present study concluded that rapamycin reduces seizures and associated cardiac damage by inhibiting mTOR activation in the zebrafish kindling model.
Collapse
Affiliation(s)
- Supriya Sharma
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Anil Kumar Rana
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Aditi Sharma
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
8
|
Zhou Y, Lin R, Lee JJ, Li D, Wang L, Li R, Yuan Y. TITE-BOIN12: A Bayesian phase I/II trial design to find the optimal biological dose with late-onset toxicity and efficacy. Stat Med 2022; 41:1918-1931. [PMID: 35098585 PMCID: PMC9199061 DOI: 10.1002/sim.9337] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 12/19/2021] [Accepted: 01/09/2022] [Indexed: 12/17/2022]
Abstract
In the era of immunotherapies and targeted therapies, the focus of early phase clinical trials has shifted from finding the maximum tolerated dose to identifying the optimal biological dose (OBD), which maximizes the toxicity-efficacy trade-off. One major impediment to using adaptive designs to find OBD is that efficacy or/and toxicity are often late-onset, hampering the designs' real-time decision rules for treating new patients. To address this issue, we propose the model-assisted TITE-BOIN12 design to find OBD with late-onset toxicity and efficacy. As an extension of the BOIN12 design, the TITE-BOIN12 design also uses utility to quantify the toxicity-efficacy trade-off. We consider two approaches, Bayesian data augmentation and an approximated likelihood method, to enable real-time decision making when some patients' toxicity and efficacy outcomes are pending. Extensive simulations show that, compared to some existing designs, TITE-BOIN12 significantly shortens the trial duration while having comparable or higher accuracy to identify OBD and a lower risk of overdosing patients. To facilitate the use of the TITE-BOIN12 design, we develop a user-friendly software freely available at http://www.trialdesign.org.
Collapse
Affiliation(s)
- Yanhong Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - J. Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Daniel Li
- Juno Therapeutics, a Bristol-Myers Squibb Company, WA, USA
| | - Li Wang
- Org Division, AbbVie Inc., IL, USA
| | - Ruobing Li
- The Center for Drug Evaluation, The National Medical Products Administration, Beijing, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, USA
| |
Collapse
|
9
|
Chou CL, Juan SH, Li CH, Chen HH, Kao CC, Chen LY, Chien LN, Fang TC. Association Between DPP-4 Inhibitors and Events of Colorectal and Liver Cancers in Patients With Diabetes Receiving Second-Line Agents: A Nested Case-Control Study. Front Oncol 2022; 12:840142. [PMID: 35600378 PMCID: PMC9120816 DOI: 10.3389/fonc.2022.840142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/12/2022] [Indexed: 12/20/2022] Open
Abstract
ObjectivePlasma dipeptidyl peptidase-4 (DPP4) levels were significantly lower in patients with colorectal and liver cancers, and animal studies also showed DPP4 inhibitors (DPP4is) have procarcinogenic effects in colorectal cancer. Until now, whether DPP4is therapy affects the progression of liver cancer and colorectal cancer in patients with T2DM has not been well investigated. We investigated the association between cumulative defined daily dose (cDDD) of DPP4is exposure and risks of liver and colorectal cancers in patients with type 2 diabetes.Materials and MethodsWe identified 268,520 patients with diabetes receiving DPP4is as second-line agents between March 1, 2009, and December 31, 2013, from Taiwan’s National Health Insurance Research Database, Taiwan Cancer Registry, and National Death Registry of Taiwan. The amount of DPP4is were divided into three groups (low, medium, and high) based on the interquartile range of the cDDD of the DPP4is.ResultsThe data showed that the low cDDD of DPP-4is was associated with a reducing risk of colorectal cancer [adjusted odds ratio (OR), 0.49; 95% CI, 0.32–0.75; P=0.001]. However, the high cDDD of DPP-4is was associated with an increasing risk of colorectal cancer (adjusted OR, 1.86; 95% CI, 1.32–2.61; P<0.001). No association between DPP4is use and liver cancer risk was observed.ConclusionsThis nested case study revealed a J-shaped association between the cDDD of DPP-4is and colorectal cancer risk, but not liver cancer risk. Therefore, the effects of long-term DPP4is use on colorectal cancer risk warrant further study.
Collapse
Affiliation(s)
- Chu-Lin Chou
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hao Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsi-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chin Kao
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Li-Ying Chen
- Health Data Analytics and Statistics Center, Office of Data Science, Taipei Medical University, Taipei, Taiwan
| | - Li-Nien Chien
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Te-Chao Fang, ; Li-Nien Chien,
| | - Te-Chao Fang
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Te-Chao Fang, ; Li-Nien Chien,
| |
Collapse
|
10
|
Jin H, Yin G. CFO: Calibration-free odds design for phase I/II clinical trials. Stat Methods Med Res 2022; 31:1051-1066. [PMID: 35238697 PMCID: PMC9527856 DOI: 10.1177/09622802221079353] [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] [Indexed: 11/16/2022]
Abstract
Recent revolution in oncology treatment has witnessed emergence and fast development of the targeted therapy and immunotherapy. In contrast to traditional cytotoxic agents, these types of treatment tend to be more tolerable and thus efficacy is of more concern. As a result, seamless phase I/II trials have gained enormous popularity, which aim to identify the optimal biological dose (OBD) rather than the maximum tolerated dose (MTD). To enhance the accuracy and robustness for identification of OBD, we develop a calibration-free odds (CFO) design. For toxicity monitoring, the CFO design casts the current dose in competition with its two neighboring doses to obtain an admissible set. For efficacy monitoring, CFO selects the dose that has the largest posterior probability to achieve the highest efficacy under the Bayesian paradigm. In contrast to most of the existing designs, the prominent merit of CFO is that its main dose-finding component is model-free and calibration-free, which can greatly ease the burden on artificial input of design parameters and thus enhance the robustness and objectivity of the design. Extensive simulation studies demonstrate that the CFO design strikes a good balance between efficiency and safety for MTD identification under phase I trials, and yields comparable or sometimes slightly better performance for OBD identification than the competing methods under phase I/II trials.
Collapse
Affiliation(s)
- Huaqing Jin
- Department of Statistics and Actuarial Science, 25809The University of Hong Kong, Pokfulam Road, Hong Kong
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, 25809The University of Hong Kong, Pokfulam Road, Hong Kong
| |
Collapse
|
11
|
Stegelmeier AA, Santry LA, Guilleman MM, Matuszewska K, Minott JA, Yates JGE, Stevens BAY, Thomas SP, Vanderkamp S, Hanada K, Pei Y, Rghei AD, van Vloten JP, Pereira M, Thompson B, Major PP, Petrik JJ, Bridle BW, Wootton SK. AAV-Vectored Expression of the Vascular Normalizing Agents 3TSR and Fc3TSR, and the Anti-Angiogenic Bevacizumab Extends Survival in a Murine Model of End-Stage Epithelial Ovarian Carcinoma. Biomedicines 2022; 10:biomedicines10020362. [PMID: 35203573 PMCID: PMC8962366 DOI: 10.3390/biomedicines10020362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Epithelial ovarian cancer is the deadliest gynecological malignancy. The lack of effective treatments highlights the need for novel therapeutic interventions. The aim of this study was to investigate whether sustained adeno-associated virus (AAV) vector-mediated expression of vascular normalizing agents 3TSR and Fc3TSR and the antiangiogenic monoclonal antibody, Bevacizumab, with or without oncolytic virus treatment would improve survival in an orthotopic syngeneic mouse model of epithelial ovarian carcinoma. AAV vectors were administered 40 days post-tumor implantation and combined with oncolytic avian orthoavulavirus-1 (AOaV-1) 20 days later, at the peak of AAV-transgene expression, to ascertain whether survival could be extended. Flow cytometry conducted on blood samples, taken at an acute time point post-AOaV-1 administration (36 h), revealed a significant increase in activated NK cells in the blood of all mice that received AOaV-1. T cell analysis revealed a significant increase in CD8+ tumor specific T cells in the blood of AAV-Bevacizumab+AOaV-1 treated mice compared to control mice 10 days post AOaV-1 administration. Immunohistochemical staining of primary tumors harvested from a subset of mice euthanized 90 days post tumor implantation, when mice typically have large primary tumors, secondary peritoneal lesions, and extensive ascites fluid production, revealed that AAV-3TSR, AAV-Fc3TSR+AOaV-1, or AAV-Bevacizumab+AOaV-1 treated mice had significantly more tumor-infiltrating CD8+ T cells than PBS controls. Despite AAV-mediated transgene expression waning faster in tumor-bearing mice than in non-tumor bearing mice, all three of the AAV therapies significantly extended survival compared to control mice; with AAV-Bevacizumab performing the best in this model. However, combining AAV therapies with a single dose of AOaV-1 did not lead to significant extensions in survival compared to AAV therapies on their own, suggesting that additional doses of AOaV-1 may be required to improve efficacy in this model. These results suggest that vectorizing anti-angiogenic and vascular normalizing agents is a viable therapeutic option that warrants further investigation, including optimizing combination therapies.
Collapse
Affiliation(s)
- Ashley A. Stegelmeier
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Lisa A. Santry
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Matthew M. Guilleman
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Kathy Matuszewska
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (K.M.); (M.P.); (J.J.P.)
| | - Jessica A. Minott
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Jacob G. E. Yates
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Brenna A. Y. Stevens
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Sylvia P. Thomas
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Sierra Vanderkamp
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Kiersten Hanada
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Amira D. Rghei
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Jacob P. van Vloten
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (K.M.); (M.P.); (J.J.P.)
| | | | - Pierre P. Major
- Juravinski Cancer Centre, 699 Concession Street, Hamilton, ON L8V 5C2, Canada;
| | - James J. Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (K.M.); (M.P.); (J.J.P.)
| | - Byram W. Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.A.S.); (L.A.S.); (M.M.G.); (J.A.M.); (J.G.E.Y.); (B.A.Y.S.); (S.P.T.); (S.V.); (K.H.); (Y.P.); (A.D.R.); (J.P.v.V.); (B.W.B.)
- Correspondence: ; Tel.: +1-519-824-4210 (ext. 54729)
| |
Collapse
|
12
|
Matuszewska K, Ten Kortenaar S, Pereira M, Santry LA, Petrik D, Lo KM, Bridle BW, Wootton SK, Lawler J, Petrik J. Addition of an Fc-IgG induces receptor clustering and increases the in vitro efficacy and in vivo anti-tumor properties of the thrombospondin-1 type I repeats (3TSR) in a mouse model of advanced stage ovarian cancer. Gynecol Oncol 2021; 164:154-169. [PMID: 34799137 DOI: 10.1016/j.ygyno.2021.11.006] [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: 08/15/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Tumor vasculature is structurally abnormal, with anatomical deformities, reduced pericyte coverage and low tissue perfusion. As a result of this vascular dysfunction, tumors are often hypoxic, which is associated with an aggressive tumor phenotype, and reduced delivery of therapeutic compounds to the tumor. We have previously shown that a peptide containing the thrombospondin-1 type I repeats (3TSR) specifically targets tumor vessels and induces vascular normalization in a mouse model of epithelial ovarian cancer (EOC). However, due to its small size, 3TSR is rapidly cleared from circulation. We now introduce a novel construct with the 3TSR peptide fused to the C-terminus of each of the two heavy chains of the Fc region of human IgG1 (Fc3TSR). We hypothesize that Fc3TSR will have greater anti-tumor activity in vitro and in vivo compared to the native compound. METHODS Fc3TSR was evaluated in vitro using proliferation and apoptosis assays to investigate differences in efficacy compared to native 3TSR. In light of the multivalency of Fc3TSR, we also investigate whether it induces greater clustering of its functional receptor, CD36. We also compare the compounds in vivo using an orthotopic, syngeneic mouse model of advanced stage EOC. The impact of the two compounds on changes to tumor vasculature morphology was also investigated. RESULTS Fc3TSR significantly decreased the viability and proliferative potential of EOC cells and endothelial cells in vitro compared to native 3TSR. High-resolution imaging followed by image correlation spectroscopy demonstrated enhanced clustering of the CD36 receptor in cells treated with Fc3TSR. This was associated with enhanced downstream signaling and greater in vitro and in vivo cellular responses. Fc3TSR induced greater vascular normalization and disease regression compared to native 3TSR in an orthotopic, syngeneic mouse model of advanced stage ovarian cancer. CONCLUSION The development of Fc3TSR which is greater in size, stable in circulation and enhances receptor activation compared to 3TSR, facilitates its translational potential as a therapy in the treatment of metastatic advanced stage ovarian cancer.
Collapse
Affiliation(s)
- Kathy Matuszewska
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, United States of America
| | - Simone Ten Kortenaar
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, United States of America
| | - Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, United States of America
| | - Lisa A Santry
- Department of Pathobiology, University of Guelph, Guelph, ON, United States of America
| | - Duncan Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, United States of America
| | - Kin-Ming Lo
- EMD Serono Research & Development Institute, Billerica, MA, United States of America
| | - Byram W Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON, United States of America
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON, United States of America
| | - Jack Lawler
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, United States of America.
| |
Collapse
|
13
|
Ali H, Khan F, Musharraf SG. Cilostazol-mediated reversion of γ-globin silencing is associated with a high level of HbF production: A potential therapeutic candidate for β-globin disorders. Biomed Pharmacother 2021; 142:112058. [PMID: 34426256 DOI: 10.1016/j.biopha.2021.112058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022] Open
Abstract
Reversal of fetal hemoglobin (HbF) silencing is an attractive therapeutic intervention for β-thalassemia and sickle cell anemia. The current study proposes the therapeutic of repurposing of cilostazol, an FDA-approved antithrombotic agent, as a promising HbF inducer. Preliminary, we report that cilostazol induced erythroid differentiation and hemoglobinization of human erythroleukemia K562 cells. The erythroid differentiation was accompanied by increased expression of γ-globin mRNA transcripts and HbF production. Cilostazol induced erythroid differentiation and HbF production, without significantly affecting proliferation and viability of hemoglobin producing cells at maximum erythroid inducing concentration. Moreover, we investigated the effect of cilostazol on human β- and γ-globin transgenes in in vivo β-YAC transgenic mice, harboring human β-locus along with β-LCR. A good in vitro correlation was found with substantial up-regulation in fetal globin mRNA; whereas, the β-globin gene expression was not significantly changed. F-cells, analysis in the peripheral blood of cilostazol-treated mice, revealed a significant increase in the F-cells population as compared with sham control groups. Together, these findings support the potential of cilostazol as an HbF inducer, which can be evaluated further to develop a new HbF inducer.
Collapse
Affiliation(s)
- Hamad Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Faisal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Syed Ghulam Musharraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
14
|
Kant R, Yang MH, Tseng CH, Yen CH, Li WY, Tyan YC, Chen M, Tzeng CC, Chen WC, You K, Wang WC, Chen YL, Chen YMA. Discovery of an Orally Efficacious MYC Inhibitor for Liver Cancer Using a GNMT-Based High-Throughput Screening System and Structure-Activity Relationship Analysis. J Med Chem 2021; 64:8992-9009. [PMID: 34132534 DOI: 10.1021/acs.jmedchem.1c00093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glycine-N-methyl transferase (GNMT) downregulation results in spontaneous hepatocellular carcinoma (HCC). Overexpression of GNMT inhibits the proliferation of liver cancer cell lines and prevents carcinogen-induced HCC, suggesting that GNMT induction is a potential approach for anti-HCC therapy. Herein, we used Huh7 GNMT promoter-driven screening to identify a GNMT inducer. Compound K78 was identified and validated for its induction of GNMT and inhibition of Huh7 cell growth. Subsequently, we employed structure-activity relationship analysis and found a potent GNMT inducer, K117. K117 inhibited Huh7 cell growth in vitro and xenograft in vivo. Oral administration of a dosage of K117 at 10 mpk (milligrams per kilogram) can inhibit Huh7 xenograft in a manner equivalent to the effect of sorafenib at a dosage of 25 mpk. A mechanistic study revealed that K117 is an MYC inhibitor. Ectopic expression of MYC using CMV promoter blocked K117-mediated MYC inhibition and GNMT induction. Overall, K117 is a potential lead compound for HCC- and MYC-dependent cancers.
Collapse
Affiliation(s)
- Rajni Kant
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Ming-Hui Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,Research Center for Natural Products and Drug Development, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wei-You Li
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Marcelo Chen
- Department of Urology, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Cherng-Chyi Tzeng
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wei-Cheng Chen
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Kaiting You
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Wen-Chieh Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
| | - Yeh-Long Chen
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Ming Arthur Chen
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| |
Collapse
|
15
|
Siddiqui H, Shafi S, Ali H, Musharraf SG. Synthesis and Erythroid Induction Activity of New Thiourea Derivatives. Med Chem 2021; 17:121-133. [PMID: 32407283 DOI: 10.2174/1573406416666200514085623] [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: 09/27/2019] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The use of medicinal agents to augment the fetal hemoglobin (HbF) accretion is an important approach for the treatment of sickle-cell anemia and β-thalassemia. HbF inducers have the potential to reduce the clinical symptoms and blood transfusion dependence in the patients of β- hemoglobinopathies. OBJECTIVE The current study was aimed to examine the erythroid induction potential of newly synthesized thiourea derivatives. METHODS Thiourea derivatives 1-27 were synthesized by using environmentally friendly methods. Compounds 3, 10 and 22 were found to be new. The structures of synthesized derivatives were deduced by using various spectroscopic techniques. These derivatives were then evaluated for their erythroid induction using the human erythroleukemic K562 cell line, as a model. The benzidine-H2O2 assay was used to evaluate erythroid induction, while HbF expression was studied through immunocytochemistry using the Anti-HbF antibody. Cytotoxicity of compounds 1-27 was also evaluated on mouse fibroblast 3T3 cell line and cancer Hela cell line using MTT assay. RESULT All the compounds (1-27) have not been reported for their erythroid induction activity previously. Compounds 1, 2, and 3 were found to be the potent erythroid inducing agents with % induction of 45± 6.9, 44± 5.9, and 41± 6.1, at 1.56, 0.78, and 0.78 μM concentrations, respectively, as compared to untreated control (12 ± 1 % induction). Furthermore, compound 1, 2, and 3 significantly induced fetal hemoglobin the expression up to 4.2-fold, 4.06-fold, and 3.52-fold, respectively, as compared to untreated control. Moreover, the compounds 1-4, 6-9, 11, 12, 15, 17, 19, 22, 23, and 25 were found to be non-cytotoxic against the 3T3 cell line. CONCLUSION This study signifies that the compounds reported here may serve as the starting point for the designing and development of new fetal hemoglobin inducers for the treatment of β- hemoglobinopathies.
Collapse
Affiliation(s)
- Hina Siddiqui
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Sarah Shafi
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Hamad Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Syed Ghulam Musharraf
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| |
Collapse
|
16
|
Kim S, Wand J, Magana-Ramirez C, Fraij J. Logistic Regression Models with Unspecified Low Dose-Response Relationships and Experimental Designs for Hormesis Studies. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2021; 41:92-109. [PMID: 32885437 DOI: 10.1111/risa.13588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/18/2020] [Accepted: 08/22/2020] [Indexed: 06/11/2023]
Abstract
Hormesis refers to a nonmonotonic (biphasic) dose-response relationship in toxicology, environmental science, and related fields. In the presence of hormesis, a low dose of a toxic agent may have a lower risk than the risk at the control dose, and the risk may increase at high doses. When the sample size is small due to practical, logistic, and ethical considerations, a parametric model may provide an efficient approach to hypothesis testing at the cost of adopting a strong assumption, which is not guaranteed to be true. In this article, we first consider alternative parameterizations based on the traditional three-parameter logistic regression. The new parameterizations attempt to provide robustness to model misspecification by allowing an unspecified dose-response relationship between the control dose and the first nonzero experimental dose. We then consider experimental designs including the uniform design (the same sample size per dose group) and the c -optimal design (minimizing the standard error of an estimator for a parameter of interest). Our simulation studies showed that (1) the c -optimal design under the traditional three-parameter logistic regression does not help reducing an inflated Type I error rate due to model misspecification, (2) it is helpful under the new parameterization with three parameters (Type I error rate is close to a fixed significance level), and (3) the new parameterization with four parameters and the c -optimal design does not reduce statistical power much while preserving the Type I error rate at a fixed significance level.
Collapse
Affiliation(s)
- Steven Kim
- Department of Mathematics and Statistics, California State University, Monterey Bay, Seaside, CA, USA
| | - Jeffrey Wand
- Department of Mathematics and Statistics, California State University, Monterey Bay, Seaside, CA, USA
| | - Christina Magana-Ramirez
- Department of Mathematics and Statistics, California State University, Monterey Bay, Seaside, CA, USA
| | - Jenel Fraij
- Department of Mathematics, Hartnell College, Salinas, CA, USA
| |
Collapse
|
17
|
Zhang Y, Zang Y. CWL: A conditional weighted likelihood method to account for the delayed joint toxicity-efficacy outcomes for phase I/II clinical trials. Stat Methods Med Res 2020; 30:892-903. [PMID: 33349166 DOI: 10.1177/0962280220979328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The delayed outcome issue is common in early phase dose-finding clinical trials. This problem becomes more intractable in phase I/II clinical trials because both toxicity and efficacy responses are subject to the delayed outcome issue. The existing methods applying for the phase I trials cannot be used directly for the phase I/II trial due to a lack of capability to model the joint toxicity-efficacy distribution. In this paper, we propose a conditional weighted likelihood (CWL) method to circumvent this issue. The key idea of the CWL method is to decompose the joint probability into the product of marginal and conditional probabilities and then weight each probability based on each patient's actual follow-up time. The CWL method makes no parametric model assumption on either the dose-response curve or the toxicity-efficacy correlation and therefore can be applied to any existing phase I/II trial design. Numerical trial applications show that the proposed CWL method yields desirable operating characteristics.
Collapse
Affiliation(s)
- Yifei Zhang
- Department of Biostatistics, Indiana University, Indianapolis, IN, USA
| | - Yong Zang
- Department of Biostatistics, Indiana University, Indianapolis, IN, USA.,Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
18
|
Signal transduction in cognitive systems: Origin and dynamics of the inverted-U/U dose-response relations. J Theor Biol 2020; 504:110377. [PMID: 32622791 DOI: 10.1016/j.jtbi.2020.110377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 11/20/2022]
Abstract
Cognitive systems are indeed physical systems, but have behavioral patterns extending far beyond familiar physical changes. These behaviors are characterized by underlying symmetries and temperature measures much different from those driving phase transitions in non-cognitive systems. We define a temperature analog dependent on a scalarized synergism between rates of internal crosstalk, external sensory information, and material supply, and show that, if the analog fits a simple concavity relation with the rate index, then the dose-response curve of cognition rate with that index will be an inverted-U. If convex, the relation will be U-shaped. The basic results can be generalized at the expense of further mathematical detail to fit a broad spectrum of empirical patterns.
Collapse
|
19
|
Afrin S, Islam MS, Patzkowsky K, Malik M, Catherino WH, Segars JH, Borahay MA. Simvastatin ameliorates altered mechanotransduction in uterine leiomyoma cells. Am J Obstet Gynecol 2020; 223:733.e1-733.e14. [PMID: 32417359 DOI: 10.1016/j.ajog.2020.05.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 04/18/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress β1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies. OBJECTIVE This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells. STUDY DESIGN This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 μM, or control) for 48 hours. Protein and mRNA levels of β1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction. RESULTS We found that simvastatin significantly reduced the protein expression of β1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in β1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1. CONCLUSION The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials.
Collapse
|
20
|
Charest G, Tippayamontri T, Shi M, Wehbe M, Anantha M, Bally M, Sanche L. Concomitant Chemoradiation Therapy with Gold Nanoparticles and Platinum Drugs Co-Encapsulated in Liposomes. Int J Mol Sci 2020; 21:E4848. [PMID: 32659905 PMCID: PMC7402338 DOI: 10.3390/ijms21144848] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
A liposomal formulation of gold nanoparticles (GNPs) and carboplatin, named LipoGold, was produced with the staggered herringbone microfluidic method. The radiosensitizing potential of LipoGold and similar concentrations of non-liposomal GNPs, carboplatin and oxaliplatin was evaluated in vitro with the human colorectal cancer cell line HCT116 in a clonogenic assay. Progression of HCT116 tumor implanted subcutaneously in NU/NU mice was monitored after an irradiation of 10 Gy combined with either LipoGold, GNPs or carboplatin injected directly into the tumor by convection-enhanced delivery. Radiosensitization by GNPs alone or carboplatin alone was observed only at high concentrations of these compounds. Furthermore, low doses of carboplatin alone or a combination of carboplatin and GNPs did not engender radiosensitization. However, the same low doses of carboplatin and GNPs administered simultaneously by encapsulation in liposomal nanocarriers (LipoGold) led to radiosensitization and efficient control of cell proliferation. Our study shows that the radiosensitizing effect of a combination of carboplatin and GNPs is remarkably more efficient when both compounds are simultaneously delivered to the tumor cells using a liposomal carrier.
Collapse
Affiliation(s)
- Gabriel Charest
- Department of Nuclear Medicine and Radiobiology and Medical Research Center, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (T.T.); (M.S.); (L.S.)
| | - Thititip Tippayamontri
- Department of Nuclear Medicine and Radiobiology and Medical Research Center, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (T.T.); (M.S.); (L.S.)
- Department of Radiological Technology and Medical Physics, Chulalongkorn University, Bangkok 10330, Thailand
| | - Minghan Shi
- Department of Nuclear Medicine and Radiobiology and Medical Research Center, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (T.T.); (M.S.); (L.S.)
| | - Mohamed Wehbe
- British Columbia Cancer Agency (BCCA), Vancouver, BC V6H 3Z6, Canada; (M.W.); (M.A.); (M.B.)
| | - Malathi Anantha
- British Columbia Cancer Agency (BCCA), Vancouver, BC V6H 3Z6, Canada; (M.W.); (M.A.); (M.B.)
| | - Marcel Bally
- British Columbia Cancer Agency (BCCA), Vancouver, BC V6H 3Z6, Canada; (M.W.); (M.A.); (M.B.)
| | - Léon Sanche
- Department of Nuclear Medicine and Radiobiology and Medical Research Center, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (T.T.); (M.S.); (L.S.)
| |
Collapse
|
21
|
Abbas R, Rossoni C, Jaki T, Paoletti X, Mozgunov P. A comparison of phase I dose-finding designs in clinical trials with monotonicity assumption violation. Clin Trials 2020; 17:522-534. [PMID: 32631095 DOI: 10.1177/1740774520932130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND/AIMS In oncology, new combined treatments make it difficult to order dose levels according to monotonically increasing toxicity. New flexible dose-finding designs that take into account uncertainty in dose levels ordering were compared with classical designs through simulations in the setting of the monotonicity assumption violation. We give recommendations for the choice of dose-finding design. METHODS Motivated by a clinical trial for patients with high-risk neuroblastoma, we considered designs that require a monotonicity assumption, the Bayesian Continual Reassessment Method, the modified Toxicity Probability Interval, the Bayesian Optimal Interval design, and designs that relax monotonicity assumption, the Bayesian Partial Ordering Continual Reassessment Method and the No Monotonicity Assumption design. We considered 15 scenarios including monotonic and non-monotonic dose-toxicity relationships among six dose levels. RESULTS The No Monotonicity Assumption and Partial Ordering Continual Reassessment Method designs were robust to the violation of the monotonicity assumption. Under non-monotonic scenarios, the No Monotonicity Assumption design selected the correct dose level more often than alternative methods on average. Under the majority of monotonic scenarios, the Partial Ordering Continual Reassessment Method selected the correct dose level more often than the No Monotonicity Assumption design. Other designs were impacted by the violation of the monotonicity assumption with a proportion of correct selections below 20% in most scenarios. Under monotonic scenarios, the highest proportions of correct selections were achieved using the Continual Reassessment Method and the Bayesian Optimal Interval design (between 52.8% and 73.1%). The costs of relaxing the monotonicity assumption by the No Monotonicity Assumption design and Partial Ordering Continual Reassessment Method were decreases in the proportions of correct selections under monotonic scenarios ranging from 5.3% to 20.7% and from 1.4% to 16.1%, respectively, compared with the best performing design and were higher proportions of patients allocated to toxic dose levels during the trial. CONCLUSIONS Innovative oncology treatments may no longer follow monotonic dose levels ordering which makes standard phase I methods fail. In such a setting, appropriate designs, as the No Monotonicity Assumption or Partial Ordering Continual Reassessment Method designs, should be used to safely determine recommended for phase II dose.
Collapse
Affiliation(s)
- Rachid Abbas
- ONCOSTAT Team CESP INSERM U1018, Univ. Paris-Saclay and Biostatistics and Epidemiology department, Gustave Roussy Cancer Center, Villejuif, France
| | - Caroline Rossoni
- ONCOSTAT Team CESP INSERM U1018, Univ. Paris-Saclay and Biostatistics and Epidemiology department, Gustave Roussy Cancer Center, Villejuif, France
| | - Thomas Jaki
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK.,MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Xavier Paoletti
- Université Versailles St Quentin & INSERM U900 STAMPM, Institut Curie, Paris, France
| | - Pavel Mozgunov
- Department of Mathematics and Statistics, Lancaster University, Lancaster, UK
| |
Collapse
|
22
|
Drevinek P, Pressler T, Cipolli M, De Boeck K, Schwarz C, Bouisset F, Boff M, Henig N, Paquette-Lamontagne N, Montgomery S, Perquin J, Tomkinson N, den Hollander W, Elborn JS. Antisense oligonucleotide eluforsen is safe and improves respiratory symptoms in F508DEL cystic fibrosis. J Cyst Fibros 2020; 19:99-107. [DOI: 10.1016/j.jcf.2019.05.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/05/2019] [Accepted: 05/20/2019] [Indexed: 10/26/2022]
|
23
|
Zhou Y, Lee JJ, Yuan Y. A utility-based Bayesian optimal interval (U-BOIN) phase I/II design to identify the optimal biological dose for targeted and immune therapies. Stat Med 2019; 38:5299-5316. [PMID: 31621952 DOI: 10.1002/sim.8361] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 05/15/2019] [Accepted: 08/12/2019] [Indexed: 11/08/2022]
Abstract
In the era of targeted therapy and immunotherapy, the objective of dose finding is often to identify the optimal biological dose (OBD), rather than the maximum tolerated dose. We develop a utility-based Bayesian optimal interval (U-BOIN) phase I/II design to find the OBD. We jointly model toxicity and efficacy using a multinomial-Dirichlet model, and employ a utility function to measure dose risk-benefit trade-off. The U-BOIN design consists of two seamless stages. In stage I, the Bayesian optimal interval design is used to quickly explore the dose space and collect preliminary toxicity and efficacy data. In stage II, we continuously update the posterior estimate of the utility for each dose after each cohort, using accumulating efficacy and toxicity from both stages I and II, and then use the posterior estimate to direct the dose assignment and selection. Compared to existing phase I/II designs, one prominent advantage of the U-BOIN design is its simplicity for implementation. Once the trial is designed, it can be easily applied using predetermined decision tables, without complex model fitting and estimation. Our simulation study shows that, despite its simplicity, the U-BOIN design is robust and has high accuracy to identify the OBD. We extend the design to accommodate delayed efficacy by leveraging the short-term endpoint (eg, immune activity or other biological activity of targeted agents), and using it to predict the delayed efficacy outcome to facilitate real-time decision making. A user-friendly software to implement the U-BOIN is freely available at www.trialdesign.org.
Collapse
Affiliation(s)
- Yanhong Zhou
- Quantitative Science, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
24
|
SREBP1-dependent de novo fatty acid synthesis gene expression is elevated in malignant melanoma and represents a cellular survival trait. Sci Rep 2019; 9:10369. [PMID: 31316083 PMCID: PMC6637239 DOI: 10.1038/s41598-019-46594-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 07/01/2019] [Indexed: 02/07/2023] Open
Abstract
de novo fatty acid biosynthesis (DNFA) is a hallmark adaptation of many cancers that supports survival, proliferation, and metastasis. Here we elucidate previously unexplored aspects of transcription regulation and clinical relevance of DNFA in cancers. We show that elevated expression of DNFA genes is characteristic of many tumor types and correlates with poor prognosis, especially in melanomas. Elevated DNFA gene expression depends on the SREBP1 transcription factor in multiple melanoma cell lines. SREBP1 predominantly binds to the transcription start sites of DNFA genes, regulating their expression by recruiting RNA polymerase II to promoters for productive transcription elongation. We find that SREBP1-regulated DNFA represents a survival trait in melanoma cells, regardless of proliferative state and oncogenic mutation status. Indeed, malignant melanoma cells exhibit elevated DNFA gene expression after the BRAF/MEK signaling pathway is blocked (e.g. by BRAF inhibitors), and DNFA expression remains higher in melanoma cells resistant to vemurafenib treatment than in untreated cells. Accordingly, DNFA pathway inhibition, whether by direct targeting of SREBP1 with antisense oligonucleotides, or through combinatorial effects of multiple DNFA enzyme inhibitors, exerts potent cytotoxic effects on both BRAFi-sensitive and -resistant melanoma cells. Altogether, these results implicate SREBP1 and DNFA enzymes as enticing therapeutic targets in melanomas.
Collapse
|
25
|
Yan D, Tait C, Wages NA, Kindwall-Keller T, Dressler EV. Generalization of the time-to-event continual reassessment method to bivariate outcomes. J Biopharm Stat 2019; 29:635-647. [PMID: 31264936 DOI: 10.1080/10543406.2019.1634087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This article considers the problem of designing Phase I-II clinical trials with delayed toxicity and efficacy outcomes. The proposed design is motivated by a Phase I-II study evaluating all-trans retinoic acid (ATRA) in combination with a fixed dose of daratumumab in the treatment of relapsed or refractory multiple myeloma. The primary objective of the study is to identify a dose that maximizes efficacy and has an acceptable level of toxicity. The toxicity endpoint is observed in one cycle of therapy (i.e., 4 weeks) while the efficacy endpoint is assessed after 8 weeks of treatment. The difference in endpoint observation windows causes logistical challenges in conducting the trial, since it is not practical to wait until both outcomes for each patient have been fully observed before sequentially assigning the dose of a newly eligible patient. In order to avoid delays in treatment for newly enrolled patients and to accelerate trial progress, we generalize the time-to-event continual reassessment method (TITE-CRM) to bivariate outcomes. Simulation studies are conducted to evaluate the proposed method, and we found that the proposed design is able to accurately select doses that maximize efficacy and have acceptable toxicity, while using all available information in allocating patients at the time of dose assignment. We compare the proposed methodology to two existing methods in the area.
Collapse
Affiliation(s)
- Donglin Yan
- a Department of Biostatistics, College of Public Health, University of Kentucky , Lexington , Kentucky , USA
| | - Christopher Tait
- b Department of Biostatistics, PRA Health Sciences , Charlottesville , Virginia , USA
| | - Nolan A Wages
- c Department of Public Health Sciences, University of Virginia , Charlottesville , Virginia , USA
| | - Tamila Kindwall-Keller
- d Division of Hematology/Oncology, University of Virginia Health System , Charlottesville , Virginia , USA
| | - Emily V Dressler
- e Department of Biostatistical Sciences, Wake Forest School of Medicine , Winston-Salem , North Carolina , USA
| |
Collapse
|
26
|
Theile D, Cho WC. Pharmacodynamic monitoring using biomarkers to individualize pharmacotherapy. Biomark Med 2019; 13:393-408. [DOI: 10.2217/bmm-2018-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drug doses are often titrated upon their clinical effects (e.g., blood pressure). Unfortunately, for many drugs there is no direct, clinical read-out to estimate dose adequateness. Alternatively, drug dosing is based on the maximum tolerated dose approach or therapeutic drug monitoring. However, the concentration-response curves may be flattened or bell-shaped as suggested for some ‘biologicals’. Together, these aspects raise the question why drug dosing is not individualized by pharmacodynamic monitoring. Evaluating the effects of drugs at their pharmacological target or meaningful biomarkers might indicate nonresponders, objectively quantify the maximum molecular effect and thus restrict overdose and underdosing. This review outlines the theory and biological or technical prerequisites for biomarker-based pharmacodynamic monitoring, and highlights selected examples from different fields of clinical medicine.
Collapse
Affiliation(s)
- Dirk Theile
- Department of Clinical Pharmacology & Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, Heidelberg 69120, Germany
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Kowloon, Hong Kong
| |
Collapse
|
27
|
Atif F, Yousuf S, Espinosa-Garcia C, Sergeeva E, Stein DG. Progesterone Treatment Attenuates Glycolytic Metabolism and Induces Senescence in Glioblastoma. Sci Rep 2019; 9:988. [PMID: 30700763 PMCID: PMC6353890 DOI: 10.1038/s41598-018-37399-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022] Open
Abstract
We examined the effect of progesterone treatments on glycolytic metabolism and senescence as possible mechanisms in controlling the growth of glioblastoma multiforme (GBM). In an orthotopic mouse model, after tumor establishment, athymic nude mice received treatment with progesterone or vehicle for 40 days. Compared to controls, high-dose progesterone administration produced a significant reduction in tumor size (~47%) and an increased survival rate (~43%) without any demonstrable toxicity to peripheral organs (liver, kidney). This was accompanied by a significant improvement in spontaneous locomotor activity and reduced anxiety-like behavior. In a follow-up in vitro study of U87MG-luc, U87dEGFR and U118MG tumor cells, we observed that high-dose progesterone inhibited expression of Glut1, which facilitated glucose transport into the cytoplasm; glyceraldehyde 3-phosphate dehydrogenase (GAPDH; a glycolysis enzyme); ATP levels; and cytoplasmic FoxO1 and Phospho-FoxO1, both of which control glycolytic metabolism through upstream PI3K/Akt/mTOR signaling in GBM. In addition, progesterone administration attenuated EGFR/PI3K/Akt/mTOR signaling, which is highly activated in grade IV GBM. High-dose progesterone also induced senescence in GBM as evidenced by changes in cell morphology and β-galactocidase accumulation. In conclusion, progesterone inhibits the modulators of glycolytic metabolism and induces premature senescence in GBM cells and this can help to reduce/slow tumor progression.
Collapse
Affiliation(s)
- Fahim Atif
- Brain Research Laboratory, Department of Emergency Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| | - Seema Yousuf
- Brain Research Laboratory, Department of Emergency Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Claudia Espinosa-Garcia
- Brain Research Laboratory, Department of Emergency Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elena Sergeeva
- Brain Research Laboratory, Department of Emergency Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Donald G Stein
- Brain Research Laboratory, Department of Emergency Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
28
|
Tykhomyrov AA, Nedzvetsky VS, Zabida AA, Ağca CA, Kuryata OV. l-Arginine treatment improves angiogenic response and reduces matrix metalloproteinase activity in chronic heart failure patients with coronary artery disease. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2018.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
29
|
Vaniotis G, Moffett S, Sulea T, Wang N, Elahi SM, Lessard E, Baardsnes J, Perrino S, Durocher Y, Frystyk J, Massie B, Brodt P. Enhanced anti-metastatic bioactivity of an IGF-TRAP re-engineered to improve physicochemical properties. Sci Rep 2018; 8:17361. [PMID: 30478273 PMCID: PMC6255772 DOI: 10.1038/s41598-018-35407-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/07/2018] [Indexed: 01/22/2023] Open
Abstract
The insulin-like growth factor (IGF) axis has been implicated in the progression of malignant disease and identified as a clinically important therapeutic target. Several IGF-1 receptor (IGF-1R) targeting drugs including humanized monoclonal antibodies have advanced to phase II/III clinical trials, but to date, have not progressed to clinical use, due, at least in part, to interference with insulin receptor signalling. We previously reported on the production of a soluble fusion protein consisting of the extracellular domain of human IGF-1R fused to the Fc portion of human IgG1 (first generation IGF-TRAP) that bound human IGF-1 and IGF-2 with a 3 log higher affinity than insulin. We showed that the IGF-TRAP had potent anti-cancer activity in several pre-clinical models of aggressive carcinomas. Here we report on the re-engineering of the IGF-TRAP with the aim of improving physicochemical properties and suitability for clinical applications. We show that cysteine-serine substitutions in the Fc hinge region of IGF-TRAP eliminated high-molecular-weight oligomerized species, while a further addition of a flexible linker, not only improved the pharmacokinetic profile, but also enhanced the therapeutic profile of the IGF-TRAP, as evaluated in an experimental colon carcinoma metastasis model. Dose-response profiles of the modified IGF-TRAPs correlated with their bio-availability profiles, as measured by the IGF kinase-receptor-activation (KIRA) assay, providing a novel, surrogate biomarker for drug efficacy. This study provides a compelling example of structure-based re-engineering of Fc-fusion-based biologics for better manufacturability that also significantly improved pharmacological parameters. It identifies the re-engineered IGF-TRAP as a potent anti-cancer therapeutic.
Collapse
Affiliation(s)
- George Vaniotis
- Department of Surgery, McGill University, Montreal Quebec, Canada
| | - Serge Moffett
- Department of Surgery, McGill University, Montreal Quebec, Canada
| | - Traian Sulea
- Institute of Parasitology, McGill University, Montreal Quebec, Canada
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Ni Wang
- Department of Surgery, McGill University, Montreal Quebec, Canada
| | - S Mehdy Elahi
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Etienne Lessard
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | | | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Jan Frystyk
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Bernard Massie
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal Quebec, Canada
| | - Pnina Brodt
- Department of Surgery, McGill University, Montreal Quebec, Canada.
- Department of Medicine, McGill University, Montreal Quebec, Canada.
- Department of Oncology, McGill University, Montreal Quebec, Canada.
- Cancer Research Program, Research Institute of the McGill University Health Center, Montreal Quebec, Canada.
| |
Collapse
|
30
|
Zhu Y, Ramasawmy R, Johnson SP, Taylor V, Gibb A, Pedley RB, Chattopadhyay N, Lythgoe MF, Golay X, Bradley D, Walker-Samuel S. Non-invasive imaging of disrupted protein homeostasis induced by proteasome inhibitor treatment using chemical exchange saturation transfer MRI. Sci Rep 2018; 8:15068. [PMID: 30305717 PMCID: PMC6180115 DOI: 10.1038/s41598-018-33549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 10/02/2018] [Indexed: 11/09/2022] Open
Abstract
Proteasome inhibitors (PIs) are now standard of care for several cancers, and noninvasive biomarkers of treatment response are critically required for early patient stratification and treatment personalization. The present study evaluated whether chemical exchange (CEST) magnetic resonance imaging (MRI) can provide measurements that can be used as the noninvasive biomarkers of proteasome inhibition, alongside diffusion MRI and relaxometry. The sensitivity of human colorectal carcinoma cells to the PI Ixazomib was assessed via in vitro and in vivo dose-response experiments. Acute in vivo response to Ixazomib was assessed at three dosing concentrations, using CEST MRI (amide, amine, hydroxyl signals), diffusion MRI (ADC) and relaxometry (T1, T2). These responses were further evaluated with the known histological markers for Ixazomib and Bradford assay ex vivo. The CEST signal from amides and amines increased in proportion to Ixazomib dose in colorectal cancer xenografts. The cell lines differed in their sensitivity to Ixazomib, which was reflected in the MRI measurements. A mild stimulation in tumor growth was observed at low Ixazomib doses. Our results identify CEST MRI as a promising method for safely and noninvasively monitoring disrupted tumor protein homeostasis induced by proteasome inhibitor treatment, and for stratifying sensitivity between tumor types.
Collapse
Affiliation(s)
- Yanan Zhu
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Rajiv Ramasawmy
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Sean Peter Johnson
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Valerie Taylor
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Alasdair Gibb
- Neuroscience, Physiology & Pharmacology, University College London, London, WC1E 6BT, United Kingdom
| | - R Barbara Pedley
- Cancer Institute, University College London, London, WC1E 6DD, United Kingdom
| | - Nibedita Chattopadhyay
- Cancer Pharmacology, Takeda Pharmaceutical International Corporation, Cambridge, MA, 02139, United States
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom
| | - Xavier Golay
- Institute of Neurology, University College London, London, WC1N 3BG, United Kingdom
| | - Daniel Bradley
- Biomedical Imaging Group, Takeda Pharmaceutical International Corporation, Cambridge, MA, 02139, United States
| | - Simon Walker-Samuel
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, United Kingdom.
| |
Collapse
|
31
|
Klement GL, Arkun K, Valik D, Roffidal T, Hashemi A, Klement C, Carmassi P, Rietman E, Slaby O, Mazanek P, Mudry P, Kovacs G, Kiss C, Norga K, Konstantinov D, André N, Slavc I, van Den Berg H, Kolenova A, Kren L, Tuma J, Skotakova J, Sterba J. Future paradigms for precision oncology. Oncotarget 2018; 7:46813-46831. [PMID: 27223079 PMCID: PMC5216837 DOI: 10.18632/oncotarget.9488] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/31/2016] [Indexed: 12/25/2022] Open
Abstract
Research has exposed cancer to be a heterogeneous disease with a high degree of inter-tumoral and intra-tumoral variability. Individual tumors have unique profiles, and these molecular signatures make the use of traditional histology-based treatments problematic. The conventional diagnostic categories, while necessary for care, thwart the use of molecular information for treatment as molecular characteristics cross tissue types. This is compounded by the struggle to keep abreast the scientific advances made in all fields of science, and by the enormous challenge to organize, cross-reference, and apply molecular data for patient benefit. In order to supplement the site-specific, histology-driven diagnosis with genomic, proteomic and metabolomics information, a paradigm shift in diagnosis and treatment of patients is required. While most physicians are open and keen to use the emerging data for therapy, even those versed in molecular therapeutics are overwhelmed with the amount of available data. It is not surprising that even though The Human Genome Project was completed thirteen years ago, our patients have not benefited from the information. Physicians cannot, and should not be asked to process the gigabytes of genomic and proteomic information on their own in order to provide patients with safe therapies. The following consensus summary identifies the needed for practice changes, proposes potential solutions to the present crisis of informational overload, suggests ways of providing physicians with the tools necessary for interpreting patient specific molecular profiles, and facilitates the implementation of quantitative precision medicine. It also provides two case studies where this approach has been used.
Collapse
Affiliation(s)
- Giannoula Lakka Klement
- Department of Pediatric Hematology/Oncology, Floating Hospital for Children at Tufts Medical Center, Boston, MA, USA.,Department of Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Knarik Arkun
- Department of Pathology, Tufts Medical Center, Boston, MA, USA
| | - Dalibor Valik
- Department of Paediatric Oncology, University Hospital Brno, Brno, Czech Republic.,Regional Center for Applied Molecular Biology, RECAMO, Brno, Czech Republic
| | - Tina Roffidal
- Department of Pediatric Hematology/Oncology, Floating Hospital for Children at Tufts Medical Center, Boston, MA, USA
| | | | | | | | - Edward Rietman
- CSTS Health Care®, Toronto, Canada.,Computer Science Department, University of Massachusetts, Amherst, MA, USA
| | - Ondrej Slaby
- Department of Paediatric Oncology, University Hospital Brno, Brno, Czech Republic.,Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Pavel Mazanek
- Department of Paediatric Oncology, University Hospital Brno, Brno, Czech Republic.,Regional Center for Applied Molecular Biology, RECAMO, Brno, Czech Republic
| | - Peter Mudry
- Department of Paediatric Oncology, University Hospital Brno, Brno, Czech Republic.,Regional Center for Applied Molecular Biology, RECAMO, Brno, Czech Republic
| | - Gabor Kovacs
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Csongor Kiss
- Department of Pediatric Hematology-Oncology, Institute of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Koen Norga
- Antwerp University Hospital, Edegem, Belgium
| | | | - Nicolas André
- Department of Pediatric Hematology and Oncology, AP-HM, Marseille, France.,UMR S_911 CRO2 Aix Marseille Université, Marseille, France
| | - Irene Slavc
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Henk van Den Berg
- Department of Pediatric Oncology, Emma Children Hospital Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Alexandra Kolenova
- Department of Pediatric Oncology, Comenius University, Bratislava, Slovakia
| | - Leos Kren
- Department of Pathology, University Hospital Brno, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiri Tuma
- Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Pediatric Surgery, University Hospital Brno, Brno, Czech Republic
| | - Jarmila Skotakova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jaroslav Sterba
- Department of Paediatric Oncology, University Hospital Brno, Brno, Czech Republic.,Faculty of Medicine, Masaryk University, Brno, Czech Republic.,ICRC St. Anna University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
32
|
Zang Y, Lee JJ, Yuan Y. Adaptive designs for identifying optimal biological dose for molecularly targeted agents. Clin Trials 2018; 11:319-327. [PMID: 24844841 DOI: 10.1177/1740774514529848] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Traditionally, the purpose of a dose-finding design in cancer is to find the maximum tolerated dose based solely on toxicity. However, for molecularly targeted agents, little toxicity may arise within the therapeutic dose range and the dose-response curves may not be monotonic. This challenges the principle that more is better, which is widely accepted for conventional chemotherapy. Methods We propose three adaptive dose-finding designs for trials evaluating molecularly targeted agents, for which the dose-response curves are unimodal or plateaued. The goal of these designs is to find the optimal biological dose, which is defined as the lowest dose with the highest rate of efficacy while safe. The first proposed design is parametric and assumes a logistic dose-efficacy curve for dose finding, the second design is nonparametric and uses the isotonic regression to identify the optimal biological dose, and the third design has the spirit of a 'semiparametric' approach by assuming a logistic model only locally around the current dose. Results We conducted extensive simulation studies to investigate the operating characteristics of the proposed designs. Simulation studies show that the nonparametric and semiparametric designs have good operating characteristics for finding the optimal biological dose. Limitations The proposed designs assume a binary endpoint. Extension of the proposed designs to ordinal and time-to-event endpoints is worth further investigation. Conclusion Among the three proposed designs, the nonparametric and semiparametric designs yield consistently good operating characteristics and thus are recommended for practical use. The software to implement these two designs is available for free download at http://odin.mdacc.tmc.edu/~yyuan/ .
Collapse
Affiliation(s)
- Yong Zang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
33
|
Jivan R, Peres J, Damelin LH, Wadee R, Veale RB, Prince S, Mavri-Damelin D. Disulfiram with or without metformin inhibits oesophageal squamous cell carcinoma in vivo. Cancer Lett 2017; 417:1-10. [PMID: 29274360 DOI: 10.1016/j.canlet.2017.12.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/30/2017] [Accepted: 12/15/2017] [Indexed: 12/18/2022]
Abstract
Oesophageal squamous cell carcinoma (OSCC) is highly prevalent in developing countries but there has been little recent progress into efficacious yet affordable treatment strategies. Drug repurposing is one attractive approach for cancer therapy. Disulfiram (DSF), used to treat alcoholism, inhibits cancer growth and we previously found that DSF perturbs protein degradation/turnover pathways in vitro. This was enhanced by combining DSF with the anti-diabetic drug metformin (Met). Here, we investigated DSF with/without Met, against OSCC in vivo. Nude mice injected subcutaneously with the human OSCC cell line WHCO1, were treated with 30 mg/kg or 50 mg/kg DSF three times per week and with/without Met, for 21 days. DSF and DSF/Met-treated animals had significantly smaller tumours compared to untreated, vehicle and positive control cisplatin-treated groups. This effect for DSF was independent of copper, with no significant accumulation of copper in tumours, together with maintained proteasome activity. However, increases in total ubiquitinated proteins, LC3B-II, LAMP1 and p62 in DSF and DSF/Met groups, indicate that autophagy is inhibited. These findings show that DSF and DSF/Met significantly impede OSCC tumour growth in vivo and offer prospective alternative chemotherapy approaches for OSCC.
Collapse
Affiliation(s)
- Rupal Jivan
- The School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Private Bag X3, WITS 2050, South Africa
| | - Jade Peres
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Leonard Howard Damelin
- The School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; Cell Biology Group, Centre for HIV and STI's, National Institute for Communicable Diseases (NHLS), Private Bag X4, Sandringham, Johannesburg 2131, South Africa
| | - Reubina Wadee
- Division of Anatomical Pathology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa
| | - Robin Bruce Veale
- The School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Private Bag X3, WITS 2050, South Africa
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town 7925, South Africa
| | - Demetra Mavri-Damelin
- The School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, Private Bag X3, WITS 2050, South Africa.
| |
Collapse
|
34
|
Liu F, Walters SJ, Julious SA. Design considerations and analysis planning of a phase 2a proof of concept study in rheumatoid arthritis in the presence of possible non-monotonicity. BMC Med Res Methodol 2017; 17:149. [PMID: 28969588 PMCID: PMC5625783 DOI: 10.1186/s12874-017-0416-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/31/2017] [Indexed: 05/10/2023] Open
Abstract
Background It is important to quantify the dose response for a drug in phase 2a clinical trials so the optimal doses can then be selected for subsequent late phase trials. In a phase 2a clinical trial of new lead drug being developed for the treatment of rheumatoid arthritis (RA), a U-shaped dose response curve was observed. In the light of this result further research was undertaken to design an efficient phase 2a proof of concept (PoC) trial for a follow-on compound using the lessons learnt from the lead compound. Methods The planned analysis for the Phase 2a trial for GSK123456 was a Bayesian Emax model which assumes the dose-response relationship follows a monotonic sigmoid “S” shaped curve. This model was found to be suboptimal to model the U-shaped dose response observed in the data from this trial and alternatives approaches were needed to be considered for the next compound for which a Normal dynamic linear model (NDLM) is proposed. This paper compares the statistical properties of the Bayesian Emax model and NDLM model and both models are evaluated using simulation in the context of adaptive Phase 2a PoC design under a variety of assumed dose response curves: linear, Emax model, U-shaped model, and flat response. Results It is shown that the NDLM method is flexible and can handle a wide variety of dose-responses, including monotonic and non-monotonic relationships. In comparison to the NDLM model the Emax model excelled with higher probability of selecting ED90 and smaller average sample size, when the true dose response followed Emax like curve. In addition, the type I error, probability of incorrectly concluding a drug may work when it does not, is inflated with the Bayesian NDLM model in all scenarios which would represent a development risk to pharmaceutical company. The bias, which is the difference between the estimated effect from the Emax and NDLM models and the simulated value, is comparable if the true dose response follows a placebo like curve, an Emax like curve, or log linear shape curve under fixed dose allocation, no adaptive allocation, half adaptive and adaptive scenarios. The bias though is significantly increased for the Emax model if the true dose response follows a U-shaped curve. Conclusions In most cases the Bayesian Emax model works effectively and efficiently, with low bias and good probability of success in case of monotonic dose response. However, if there is a belief that the dose response could be non-monotonic then the NDLM is the superior model to assess the dose response. Electronic supplementary material The online version of this article (10.1186/s12874-017-0416-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Feng Liu
- GlaxoSmithKline, Inc, 1250 South Collegeville Road, PO Box 5089, Collegeville, PA, 19426-0989, USA. .,Medical Statistics Group, University of Sheffield, Sheffield, UK.
| | | | - Steven A Julious
- Medical Statistics Group, University of Sheffield, Sheffield, UK
| |
Collapse
|
35
|
Green DS, Young HA, Valencia JC. Current prospects of type II interferon γ signaling and autoimmunity. J Biol Chem 2017; 292:13925-13933. [PMID: 28652404 PMCID: PMC5572907 DOI: 10.1074/jbc.r116.774745] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interferon γ (IFNγ) is a pleiotropic protein secreted by immune cells. IFNγ signals through the IFNγ receptor, a protein complex that mediates downstream signaling events. Studies into IFNγ signaling have provided insight into the general concepts of receptor signaling, receptor internalization, regulation of distinct signaling pathways, and transcriptional regulation. Although IFNγ is the central mediator of the adaptive immune response to pathogens, it has been shown to be involved in several non-infectious physiological processes. This review will provide an introduction into IFNγ signaling biology and the functional roles of IFNγ in the autoimmune response.
Collapse
Affiliation(s)
- Daniel S Green
- From the Women's Malignancy Branch, Translational Genomics Section, Center for Cancer, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Howard A Young
- Cancer and Inflammation Program, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702-1201.
| | - Julio C Valencia
- Cancer and Inflammation Program, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702-1201.
| |
Collapse
|
36
|
Lin R, Yin G. STEIN: A simple toxicity and efficacy interval design for seamless phase I/II clinical trials. Stat Med 2017; 36:4106-4120. [DOI: 10.1002/sim.7428] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Ruitao Lin
- Department of Biostatistics; University of Washington; Seattle 98195 Washington U.S.A
| | - Guosheng Yin
- Department of Statistics and Actuarial Science; The University of Hong Kong; Pokfulam Road Hong Kong
| |
Collapse
|
37
|
Marcinczyk M, Elmashhady H, Talovic M, Dunn A, Bugis F, Garg K. Laminin-111 enriched fibrin hydrogels for skeletal muscle regeneration. Biomaterials 2017; 141:233-242. [PMID: 28697464 DOI: 10.1016/j.biomaterials.2017.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 12/27/2022]
Abstract
Laminin (LM)-111 supplementation has improved muscle regeneration in several models of disease and injury. This study investigated a novel hydrogel composed of fibrinogen and LM-111. Increasing LM-111 concentration (50-450 μg/mL) in fibrin hydrogels resulted in highly fibrous scaffolds with progressively thinner interlaced fibers. Rheological testing showed that all hydrogels had viscoelastic behavior and the Young's modulus ranged from 2-6KPa. C2C12 myobalsts showed a significant increase in VEGF production and decrease in IL-6 production on LM-111 enriched fibrin hydrogels as compared to pure fibrin hydrogels on day 4. Western blotting results showed a significant increase in MyoD and desmin protein quantity but a significant decrease in myogenin protein quantity in myoblasts cultured on the LM-111 (450 μg/mL) enriched fibrin hydrogel. Combined application of electromechanical stimulation significantly enhanced the production of VEGF and IGF-1 from myoblast seeded fibrin-LM-111 hydrogels. Taken together, these observations offer an important first step toward optimizing a tissue engineered constructs for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, USA
| | - Hady Elmashhady
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, USA
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, USA
| | - Faiz Bugis
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, USA
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, USA.
| |
Collapse
|
38
|
Stein DG, Sayeed I, Espinosa-Garcia C, Atif F, Sergeeva EG. Goldstein et al.'s Secondary Analysis of Progesterone Clinical Trial for Traumatic Brain Injury Can Only Reflect the Same Trial Design Flaws: A Response to “Very Early Administration of Progesterone Does Not Improve Neuropsychological Outcomes in Subjects with Moderate to Severe Traumatic Brain Injury”. J Neurotrauma 2017; 34:2192-2193. [DOI: 10.1089/neu.2016.4949] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Donald G. Stein
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | | | - Fahim Atif
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | - Elena G. Sergeeva
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
39
|
Wang H, Sim MK, Loke WK, Chinnathambi A, Alharbi SA, Tang FR, Sethi G. Potential Protective Effects of Ursolic Acid against Gamma Irradiation-Induced Damage Are Mediated through the Modulation of Diverse Inflammatory Mediators. Front Pharmacol 2017; 8:352. [PMID: 28670276 PMCID: PMC5472704 DOI: 10.3389/fphar.2017.00352] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023] Open
Abstract
This study was aimed to evaluate the possible protective effects of ursolic acid (UA) against gamma radiation induced damage both in vitro as well as in vivo. It was observed that the exposure to gamma radiation dose- and time-dependently caused a significant decrease in the cell viability, while the treatment of UA attenuated this cytotoxicity. The production of free radicals including reactive oxygen species (ROS) and NO increased significantly post-irradiation and further induced lipid peroxidation and oxidative DNA damage in cells. These deleterious effects could also be effectively blocked by UA treatment. In addition, UA also reversed gamma irradiation induced inflammatory responses, as indicated by the decreased production of TNF-α, IL-6, and IL-1β. NF-κB signaling pathway has been reported to be a key mediator involved in gamma radiation-induced cellular damage. Our results further demonstrated that gamma radiation dose- and time-dependently enhanced NF-κB DNA binding activity, which was significantly attenuated upon UA treatment. The post-irradiation increase in the expression of both phospho-p65, and phospho-IκBα was also blocked by UA. Moreover, the treatment of UA was found to significantly prolong overall survival in mice exposed to whole body gamma irradiation, and reduce the excessive inflammatory responses. Given its radioprotective efficacy as described here, UA as an antioxidant and NF-κB pathway blocker, may function as an important pharmacological agent in protecting against gamma irradiation-induced injury.
Collapse
Affiliation(s)
- Hong Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
- Singapore Nuclear Research and Safety Initiative, National University of SingaporeSingapore, Singapore
| | - Meng-Kwoon Sim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Weng Keong Loke
- Agent Diagnostic and Therapeutic Laboratory, Defence and Environmental Research Institute, DSO National LaboratoriesSingapore, Singapore
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh, Saudi Arabia
| | - Feng Ru Tang
- Singapore Nuclear Research and Safety Initiative, National University of SingaporeSingapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh, Saudi Arabia
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, PerthWA, Australia
| |
Collapse
|
40
|
Stein DG. Lost and found: what we have learned from the progesterone for traumatic brain injury trials. FUTURE NEUROLOGY 2017. [DOI: 10.2217/fnl-2016-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A short opinion piece updating “Lost in translation: understanding the failure of the progesterone/traumatic brain injury Phase III trials”
Collapse
Affiliation(s)
- Donald G Stein
- Department of Emergency Medicine, Emory University, 1365 B Clifton Rd NE, Suite 5100, Atlanta, GA 30322, USA
| |
Collapse
|
41
|
Wang Y, Zhou Z, Wang W, Liu M, Bao Y. Differential effects of sulforaphane in regulation of angiogenesis in a co-culture model of endothelial cells and pericytes. Oncol Rep 2017; 37:2905-2912. [DOI: 10.3892/or.2017.5565] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/30/2017] [Indexed: 11/06/2022] Open
|
42
|
Mudry P, Slaby O, Neradil J, Soukalova J, Melicharkova K, Rohleder O, Jezova M, Seehofnerova A, Michu E, Veselska R, Sterba J. Case report: rapid and durable response to PDGFR targeted therapy in a child with refractory multiple infantile myofibromatosis and a heterozygous germline mutation of the PDGFRB gene. BMC Cancer 2017; 17:119. [PMID: 28183292 PMCID: PMC5301362 DOI: 10.1186/s12885-017-3115-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 02/04/2017] [Indexed: 11/23/2022] Open
Abstract
Background Infantile myofibromatosis belongs to a family of soft tissue tumors. The majority of these tumors have benign behavior but resistant and malignant courses are known, namely in tumors with visceral involvement. The standard of care is surgical resection. Observations suggest that low dose chemotherapy is beneficial. The treatment of resistant or relapsed patients with multifocal disease remains challenging. Patients that harbor an actionable mutation in the kinase domain are potential subjects for targeted tyrosine kinase inhibitor therapy. Case presentation An infant boy with inborn generalized infantile myofibromatosis that included bone, intracranial, soft tissue and visceral involvement was treated according to recent recommendations with low dose chemotherapy. The presence of a partial but temporary response led to a second line of treatment with six cycles of chemotherapy, which achieved a partial response again but was followed by severe toxicity. The generalized progression of the disease was observed later. Genetic analyses were performed and revealed a PDGFRB gene c.1681C>A missense heterozygous germline mutation, high PDGFRβ phosphokinase activity within the tumor and the heterozygous germline Slavic Nijmegen breakage syndrome 657del5 mutation in the NBN gene. Targeted treatment with sunitinib, the PDGFRβ inhibitor, plus low dose vinblastine led to an unexpected and durable response without toxicities or limitations to daily life activities. The presence of the Slavic NBN gene mutation limited standard chemotherapy dosing due to severe toxicities. Sister of the patient suffred from skull base tumor with same genotype and histology. The same targeted therapy led to similar quick and durable response. Conclusion Progressive and resistant incurable infantile myofibromatosis can be successfully treated with the new approach described herein. Detailed insights into the biology of the patient’s tumor and genome are necessary to understand the mechanisms of activity of less toxic and effective drugs except for up to date population-based chemotherapy regimens. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3115-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic.
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Jakub Neradil
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic.,Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Kotlarska 2, Brno, 611 37, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno, 656 91, Czech Republic
| | - Jana Soukalova
- Division of Medical Genetics, Department of Biology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic
| | - Kristyna Melicharkova
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno, 656 91, Czech Republic
| | - Ondrej Rohleder
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic
| | - Marta Jezova
- Department of Pathology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic
| | - Anna Seehofnerova
- Department of Pediatric Radiology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic
| | - Elleni Michu
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Renata Veselska
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic.,Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Kotlarska 2, Brno, 611 37, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno, 656 91, Czech Republic
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Cernopolni 9, Brno, 613 00, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno, 656 91, Czech Republic
| |
Collapse
|
43
|
Zhang N, Xia Y, Zou Y, Yang W, Zhang J, Zhong Z, Meng F. ATN-161 Peptide Functionalized Reversibly Cross-Linked Polymersomes Mediate Targeted Doxorubicin Delivery into Melanoma-Bearing C57BL/6 Mice. Mol Pharm 2017; 14:2538-2547. [PMID: 28005375 DOI: 10.1021/acs.molpharmaceut.6b00800] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PHSCN peptide (licensed as ATN-161) is an effective α5β1 integrin inhibitor that has advanced to phase II clinical trials to treat solid tumors. Here we developed ATN-161 functionalized self-cross-linkable and intracellularly de-cross-linkable polymersomes (ATN/SCID-Ps) for highly efficient and targeted delivery of doxorubicin hydrochloride (DOX·HCl) into B16F10 melanoma-bearing C57BL/6 mice. ATN/SCID-Ps exhibited a high loading capacity of DOX·HCl. The size of DOX-loaded ATN/SCID-Ps (DOX-ATN/SCID-Ps) decreased from 150 to 88 nm with increasing ATN surface densities from 0 to 100% (mol/mol). DOX-ATN/SCID-Ps were robust with low drug leakage under physiological conditions while quickly releasing DOX with the addition of 10 mM glutathione. MTT assay results displayed that DOX-ATN/SCID-Ps induced ATN density-dependent antitumor activity to α5β1 integrin overexpressing B16F10 melanoma cells, in which 56% ATN-161 was optimal. Flow cytometry and CLSM studies revealed significantly more efficient internalization and cytoplasmic DOX release in B16F10 cells for DOX-ATN/SCID-Ps than for DOX-SCID-Ps (nontargeting control) as well as clinically used pegylated liposomal doxorubicin (DOX-LPs). DOX-ATN/SCID-Ps displayed a long blood circulation time (elimination half-life = 4.13 h) and 4 times higher DOX accumulation in B16F10 bearing C57BL/6 mice than DOX-LPs. Interestingly, DOX-ATN/SCID-Ps exhibited a superior maximum-tolerated dose of over 100 mg DOX·HCl/kg, 10 times higher than DOX-LPs. Remarkably, DOX-ATN/SCID-Ps could significantly inhibit the growth of aggressive B16F10 melanoma with little adverse effects via either multiple or single injection of total dosage of 100 mg DOX·HCl/kg, resulting in greatly improved survival rates as compared to DOX-LPs. ATN/SCID-Ps are appealing nanovehicles for targeted chemotherapy of α5β1 integrin positive solid tumors.
Collapse
Affiliation(s)
- Ning Zhang
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Yifeng Xia
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Yan Zou
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Weijing Yang
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Jian Zhang
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| |
Collapse
|
44
|
Howard RB, Sayeed I, Stein DG. Suboptimal Dosing Parameters as Possible Factors in the Negative Phase III Clinical Trials of Progesterone for Traumatic Brain Injury. J Neurotrauma 2016; 34:1915-1918. [PMID: 26370183 PMCID: PMC5455214 DOI: 10.1089/neu.2015.4179] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
To date, outcomes for all Phase III clinical trials for traumatic brain injury (TBI) have been negative. The recent disappointing results of the Progesterone for the Treatment of Traumatic Brain Injury (ProTECT) and Study of a Neuroprotective Agent, Progesterone, in Severe Traumatic Brain Injury (SyNAPSe) Phase III trials for progesterone in TBI have triggered considerable speculation about the reasons for the negative outcomes of these two studies in particular and for those of all previous Phase III TBI clinical trials in general. Among the factors proposed to explain the ProTECT III and SyNAPSe results, the investigators themselves and others have cited: 1) the pathophysiological complexity of TBI itself; 2) issues with the quality and clinical relevance of the preclinical animal models; 3) insufficiently sensitive clinical endpoints; and 4) inappropriate clinical trial designs and strategies. This paper highlights three critical trial design factors that may have contributed substantially to the negative outcomes: 1) suboptimal doses and treatment durations in the Phase II studies; 2) the strategic decision not to perform Phase IIB studies to optimize these variables before initiating Phase III; and 3) the lack of incorporation of the preclinical and Chinese Phase II results, as well as allometric scaling principles, into the Phase III designs. Given these circumstances and the exceptional pleiotropic potential of progesterone as a TBI (and stroke) therapeutic, we are advocating a return to Phase IIB testing. We advocate the incorporation of dose and schedule optimization focused on lower doses and a longer duration of treatment, combined with the addressing of other potential trial design problems raised by the authors in the recently published trial results.
Collapse
Affiliation(s)
- Randy B Howard
- 1 Pharmacology Consultant, Drug Discovery and Development, Emory University , Atlanta, Georgia
| | - Iqbal Sayeed
- 2 Department of Emergency Medicine, Emory University , Atlanta, Georgia
| | - Donald G Stein
- 2 Department of Emergency Medicine, Emory University , Atlanta, Georgia
| |
Collapse
|
45
|
Maccari S, Buoncervello M, Rampin A, Spada M, Macchia D, Giordani L, Stati T, Bearzi C, Catalano L, Rizzi R, Gabriele L, Marano G. Biphasic effects of propranolol on tumour growth in B16F10 melanoma-bearing mice. Br J Pharmacol 2016; 174:139-149. [PMID: 27792834 DOI: 10.1111/bph.13662] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/05/2016] [Accepted: 10/20/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Propranolol is a vasoactive drug that shows antiangiogenic and antitumour activities in melanoma. However, it is unknown whether these activities are dose-dependent and whether there is a relationship between systemic vascular effects of propranolol and anti-melanoma activity. EXPERIMENTAL APPROACH Effects of increasing doses of propranolol (10, 20, 30 and 40 mg·kg-1 ·day-1 ) on tumour growth were studied in B16F10 melanoma-bearing mice. Histological and biochemical analyses were used to assess propranolol effects on angiogenesis and cancer cell proliferation. Systemic vascular resistance (SVR) was evaluated by measuring cardiac output and arterial BP. KEY RESULTS In vitro analyses revealed that B16F10 cells expressed β-adrenoceptors, but neither isoprenaline, a β-adrenoceptor agonist, nor the β-blocker propranolol affected cancer cell proliferation. In vivo studies showed that the antitumour efficacy of propranolol follows a U-shaped biphasic dose-response curve. Low doses (10 and 20 mg·kg-1 ·day-1 ) significantly inhibit tumour growth, whereas higher doses are progressively less effective. We also found that high-dose propranolol stimulates tumour arteriogenesis whereas no effect on angiogenesis was observed at any dose. Based on these data and considering that propranolol is a vasoactive drug, we hypothesized that it causes systemic vasoconstriction or vasodilation depending on the dose and thus alters tumour perfusion and growth. Consistent with this hypothesis, we found that propranolol has a biphasic effect on SVR with low and high doses producing vasoconstriction and vasodilation respectively. CONCLUSIONS AND IMPLICATIONS Propranolol inhibits melanoma growth in a U-shaped biphasic manner. A direct relationship exists between SVR and anti-melanoma activity.
Collapse
Affiliation(s)
- Sonia Maccari
- Department of Pharmacology, National Institute of Health, Rome, Italy
| | - Maria Buoncervello
- Department of Hematology, Oncology and Molecular Medicine, National Institute of Health, Rome, Italy
| | - Andrea Rampin
- Cell Biology and Neurobiology Institute, CNR, Rome, Italy
| | - Massimo Spada
- Department of Hematology, Oncology and Molecular Medicine, National Institute of Health, Rome, Italy
| | - Daniele Macchia
- Department of Hematology, Oncology and Molecular Medicine, National Institute of Health, Rome, Italy
| | - Luciana Giordani
- Department of Pharmacology, National Institute of Health, Rome, Italy
| | - Tonino Stati
- Department of Pharmacology, National Institute of Health, Rome, Italy
| | - Claudia Bearzi
- Cell Biology and Neurobiology Institute, CNR, Rome, Italy
| | | | - Roberto Rizzi
- Cell Biology and Neurobiology Institute, CNR, Rome, Italy
| | - Lucia Gabriele
- Department of Hematology, Oncology and Molecular Medicine, National Institute of Health, Rome, Italy
| | - Giuseppe Marano
- Department of Pharmacology, National Institute of Health, Rome, Italy
| |
Collapse
|
46
|
Muscella A, Vetrugno C, Biagioni F, Calabriso N, Calierno MT, Fornai F, De Pascali SA, Marsigliante S, Fanizzi FP. Antitumour and antiangiogenic activities of [Pt(O,O'-acac)(γ-acac)(DMS)] in a xenograft model of human renal cell carcinoma. Br J Pharmacol 2016; 173:2633-44. [PMID: 27351124 DOI: 10.1111/bph.13543] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE It is thought that the mechanism of action of anticancer chemotherapeutic agents is mainly due to a direct inhibition of tumour cell proliferation. In tumour specimens, the endothelial cell proliferation rate increases, suggesting that the therapeutic effects of anticancer agents could also be attributed to inhibition of tumour angiogenesis. Hence, we investigated the potential effects of [Pt(O,O'-acac)(γ-acac)(DMS)] ([Pt(DMS)]), a new platinum drug for non-genomic targets, on human renal carcinoma and compared them with those of the well-established anticancer drug, cisplatin. EXPERIMENTAL APPROACH Tumour growth, tumour cell proliferation and microvessel density were investigated in a xenograft model of renal cell carcinoma, developed by injecting Caki-1 cells into BALB/c nude mice. The antiangiogenic potential of compounds was also investigated using HUVECs. KEY RESULTS Treatment of the Caki-1 cells with cisplatin or [Pt(DMS)] resulted in a dose-dependent inhibition of cell survival, but the cytotoxicity of [Pt(DMS)] was approximately fivefold greater than that of cisplatin. [Pt(DMS)] was much more effective than cisplatin at inhibiting tumour growth, proliferation and angiogenesis in vivo, as well as migration, tube formation and MMP1, MMP2 and MMP9 secretion of endothelial cells in vitro. Whereas, cisplatin exerted a greater cytotoxic effect on HUVECs, but did not affect tube formation or the migration of endothelial cells. In addition, treatment of the xenograft mice with [Pt(DMS)] decreased VEGF, MMP1 and MMP2 expressions in tumours. CONCLUSIONS AND IMPLICATIONS The antiangiogenic and antitumour activities of [Pt(DMS)] provide a solid starting point for its validation as a suitable candidate for further pharmacological testing.
Collapse
Affiliation(s)
- A Muscella
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - C Vetrugno
- Neuropathology Unit, Institute of Experimental Neurology and Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - F Biagioni
- Laboratory of Neurobiology of Movement Disorders, Department of Molecular Pathology, I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - N Calabriso
- Laboratory of Nutrigenomics and Vascular Biology, Institute of Clinical Physiology, National Research Council, Lecce, Italy
| | - M T Calierno
- Laboratory of Neurobiology of Movement Disorders, Department of Molecular Pathology, I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - F Fornai
- Laboratory of Neurobiology of Movement Disorders, Department of Molecular Pathology, I.R.C.C.S. Neuromed, Pozzilli, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - S A De Pascali
- Laboratory of Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - S Marsigliante
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - F P Fanizzi
- Laboratory of Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| |
Collapse
|
47
|
Ahn EJ, Choi GJ, Kang H, Baek CW, Jung YH, Woo YC, Bang SR. Antinociceptive Effects of Ginsenoside Rg3 in a Rat Model of Incisional Pain. Eur Surg Res 2016; 57:211-223. [PMID: 27441690 DOI: 10.1159/000448001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/27/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Ginsenoside Rg3 is an extract of total ginseng saponins, which accounts for 4.7% of all saponins. This study aimed to identify the mechanisms of the antinociceptive effects of ginsenoside Rg3. METHODS Rats were randomly divided into six groups, which were treated with vehicle or 0.5, 1, 1.5, 2, or 4 mg/kg of ginsenoside Rg3 intraperitoneally 2 h after a plantar incision was made. To evaluate the mechanisms of antinociceptive effects, the rats were intraperitoneally injected with naloxone 5 mg/kg, atropine 1 mg/kg, yohimbine 2 mg/kg, mecamylamine 1 mg/kg, prazosin 1 mg/kg, and dexmedetomidine 5 μg/kg. Hyperalgesia produced by the plantar incision was assessed using von Frey filaments 1 day before the incision (BI) and 2 h after the plantar incision (AP); this measurement was repeated at 15, 30, 45, 60, 80, 100 and 120 min, and 24 and 48 h after the injection of ginsenoside Rg3. Serum interleukin-1β (IL-1β) and interleukin-6 (IL-6) levels were measured 1 day before incision and 120 min, 24 h, and 48 h after the injection of ginsenoside Rg3 or vehicle. RESULTS The mechanical withdrawal threshold (MWT) significantly increased in the group that received ginsenoside Rg3. The dose-MWT response showed a curvilinear, bell-shaped relationship. The maximum MWT was found with the administration of ginsenoside Rg3 at 1.5 mg/kg; MWT decreased to 2 and 4 mg/kg. Yohimbine diminished the analgesic effect of ginsenoside Rg3. Prazosin and dexmedetomidine increased the analgesic effect of ginsenoside Rg3. IL-1β and IL-6 appeared significantly lower relative to control group. CONCLUSIONS Ginsenoside Rg3 has an analgesic effect with a curvilinear dose-response relationship. Alpha 2 adrenergic receptor appeared to be related to the analgesic effect of ginsenoside Rg3. Also, the anti-inflammatory effect of ginsenoside Rg3 could be related to its analgesic effect.
Collapse
Affiliation(s)
- Eun Jin Ahn
- Department of Anesthesiology and Pain Medicine, Inje University Seoul Paik Hospital, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
48
|
Goey AKL, Chau CH, Sissung TM, Cook KM, Venzon DJ, Castro A, Ransom TR, Henrich CJ, McKee TC, McMahon JB, Grkovic T, Cadelis MM, Copp BR, Gustafson KR, Figg WD. Screening and Biological Effects of Marine Pyrroloiminoquinone Alkaloids: Potential Inhibitors of the HIF-1α/p300 Interaction. JOURNAL OF NATURAL PRODUCTS 2016; 79:1267-75. [PMID: 27140429 PMCID: PMC6323635 DOI: 10.1021/acs.jnatprod.5b00846] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Inhibition of the hypoxia-inducible factor 1α (HIF-1α) pathway by disrupting its association with the transcriptional coactivator p300 inhibits angiogenesis and tumor development. Development of HIF-1α/p300 inhibitors has been hampered by preclinical toxicity; therefore, we aimed to identify novel HIF-1α/p300 inhibitors. Using a cell-free assay designed to test compounds that block HIF-1α/p300 binding, 170 298 crude natural product extracts and prefractionated samples were screened, identifying 25 active extracts. One of these extracts, originating from the marine sponge Latrunculia sp., afforded six pyrroloiminoquinone alkaloids that were identified as positive hits (IC50 values: 1-35 μM). Luciferase assays confirmed inhibition of HIF-1α transcriptional activity by discorhabdin B (1) and its dimer (2), 3-dihydrodiscorhabdin C (3), makaluvamine F (5), discorhabdin H (8), discorhabdin L (9), and discorhabdin W (11) in HCT 116 colon cancer cells (0.1-10 μM, p < 0.05). Except for 11, all of these compounds also reduced HIF-1α transcriptional activity in LNCaP prostate cancer cells (0.1-10 μM, p < 0.05). These effects occurred at noncytotoxic concentrations (<50% cell death) under hypoxic conditions. At the downstream HIF-1α target level, compound 8 (0.5 μM) significantly decreased VEGF secretion in LNCaP cells (p < 0.05). In COLO 205 colon cancer cells no activity was shown in the luciferase or cytotoxicity assays. Pyrroloiminoquinone alkaloids are a novel class of HIF-1α inhibitors, which interrupt the protein-protein interaction between HIF-1α and p300 and consequently reduce HIF-related transcription.
Collapse
Affiliation(s)
- Andrew K. L. Goey
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Cindy H. Chau
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Tristan M. Sissung
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Kristina M. Cook
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - David J. Venzon
- Biostatistics & Data Management Section, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Amaya Castro
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Tanya R. Ransom
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Curtis J. Henrich
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702-1201, United States
| | - Tawnya C. McKee
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - James B. McMahon
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Tanja Grkovic
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Melissa M. Cadelis
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Brent R. Copp
- School of Chemical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Kirk R. Gustafson
- Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, Maryland 20892, United States
| |
Collapse
|
49
|
Melatonin Protects Human Adipose-Derived Stem Cells from Oxidative Stress and Cell Death. Arch Plast Surg 2016; 43:237-41. [PMID: 27218020 PMCID: PMC4876151 DOI: 10.5999/aps.2016.43.3.237] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/04/2016] [Accepted: 04/12/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Adipose-derived stem cells (ASCs) have applications in regenerative medicine based on their therapeutic potential to repair and regenerate diseased and damaged tissue. They are commonly subject to oxidative stress during harvest and transplantation, which has detrimental effects on their subsequent viability. By functioning as an antioxidant against free radicals, melatonin may exert cytoprotective effects on ASCs. METHODS We cultured human ASCs in the presence of varying dosages of hydrogen peroxide and/or melatonin for a period of 3 hours. Cell viability and apoptosis were determined with propidium iodide and Hoechst 33342 staining under fluorescence microscopy. RESULTS Hydrogen peroxide (1-2.5 mM) treatment resulted in an incremental increase in cell death. 2 mM hydrogen peroxide was thereafter selected as the dose for co-treatment with melatonin. Melatonin alone had no adverse effects on ASCs. Co-treatment of ASCs with melatonin in the presence of hydrogen peroxide protected ASCs from cell death in a dose-dependent manner, and afforded maximal protection at 100 µM (n=4, one-way analysis of variance P<0.001). Melatonin co-treated ASCs displayed significantly fewer apoptotic cells, as demonstrated by condensed and fragmented nuclei under fluorescence microscopy. CONCLUSIONS Melatonin possesses cytoprotective properties against oxidative stress in human ASCs and might be a useful adjunct in fat grafting and cell-assisted lipotransfer.
Collapse
|
50
|
Fernández-Trasancos Á, Guerola-Segura R, Paradela-Dobarro B, Álvarez E, García-Acuña JM, Fernández ÁL, González-Juanatey JR, Eiras S. Glucose and Inflammatory Cells Decrease Adiponectin in Epicardial Adipose Tissue Cells: Paracrine Consequences on Vascular Endothelium. J Cell Physiol 2016; 231:1015-1023. [PMID: 26406271 DOI: 10.1002/jcp.25189] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/08/2015] [Indexed: 11/25/2024]
Abstract
Epicardial adipose tissue (EAT) is a source of energy for heart that expresses the insulin-sensitizer, anti-inflammatory and anti-atherogenic protein, adiponectin. But, in coronary artery disease, adiponectin production declines. Our objective was to determine its regulation by glucose and inflammation in stromal cells from EAT and subcutaneous adipose tissue (SAT) and its paracrine effect on endothelial cells. Stromal cells of EAT and SAT were obtained from patients who underwent cardiac surgery. Adipogenesis was induced at 117, 200, or 295 mg/dl glucose, with or without macrophage-conditioned medium (MCM). Expression of adiponectin, GLUT-4 and the insulin receptor was analyzed by real-time PCR. The paracrine effect of stromal cells was determined in co-cultures with endothelial cells, by exposing them to high glucose and/or MCM, and, additionally, to leukocyte-conditioned medium from patients with myocardial infarction. The endothelial response was determined by analyzing vascular adhesion molecule expression. Our results showed a U-shaped dose-response curve of glucose on adiponectin in EAT, but not in SAT stromal cells. Conversely, MCM reduced the adipogenesis-induced adiponectin expression of EAT stromal cells. The presence of EAT stromal increased the inflammatory molecules of endothelial cells. This deleterious effect was emphasized in the presence of inflammatory cell-conditioned medium from patients with myocardial infarction. Thus, high glucose and inflammatory cells reduced adipogenesis-induced adiponectin expression of EAT stromal cells, which induced an inflammatory paracrine process in endothelial cells. This inflammatory effect was lower in presence of mature adipocytes, producers of adiponectin. These results contribute to understanding the role of EAT dysfunction on coronary atherosclerosis progression.
Collapse
Affiliation(s)
- Ángel Fernández-Trasancos
- Cardiology Group, Health Research Institute, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Raquel Guerola-Segura
- Department of Cardiology, Coronary Care Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Beatriz Paradela-Dobarro
- Cardiology Group, Health Research Institute, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ezequiel Álvarez
- Cardiology Group, Health Research Institute, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - José María García-Acuña
- Cardiology Group, Health Research Institute, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Cardiology, Coronary Care Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel Luis Fernández
- Department of Heart Surgery, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cardiology Group, Health Research Institute, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Cardiology, Coronary Care Unit, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Sonia Eiras
- Cardiology Group, Health Research Institute, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|