1
|
Zahoor I, Pan G, Cerghet M, Elbayoumi T, Mao-Draayer Y, Giri S, Palaniyandi SS. Current understanding of cardiovascular autonomic dysfunction in multiple sclerosis. Heliyon 2024; 10:e35753. [PMID: 39170118 PMCID: PMC11337049 DOI: 10.1016/j.heliyon.2024.e35753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Autoimmune diseases, including multiple sclerosis (MS), are proven to increase the likelihood of developing cardiovascular disease (CVD) due to a robust systemic immune response and inflammation. MS can lead to cardiovascular abnormalities that are related to autonomic nervous system dysfunction by causing inflammatory lesions surrounding tracts of the autonomic nervous system in the brain and spinal cord. CVD in MS patients can affect an already damaged brain, thus worsening the disease course by causing brain atrophy and white matter disease. Currently, the true prevalence of cardiovascular dysfunction and associated death rates in patients with MS are mostly unknown and inconsistent. Treating vascular risk factors is recommended to improve the management of this disease. This review provides an updated summary of CVD prevalence in patients with MS, emphasizing the need for more preclinical studies using animal models to understand the pathogenesis of MS better. However, no distinct studies exist that explore the temporal effects and etiopathogenesis of immune/inflammatory cells on cardiac damage and dysfunction associated with MS, particularly in the cardiac myocardium. To this end, a thorough investigation into the clinical presentation and underlying mechanisms of CVD must be conducted in patients with MS and preclinical animal models. Additionally, clinicians should monitor for cardiovascular complications while prescribing medications to MS patients, as some MS drugs cause severe CVD.
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, MI, USA
| | - Guodong Pan
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
| | - Mirela Cerghet
- Department of Neurology, Henry Ford Health, Detroit, MI, USA
| | - Tamer Elbayoumi
- Department of Pharmaceutical Sciences, College of Pharmacy, Midwestern University, Glendale, AZ, USA
| | - Yang Mao-Draayer
- Multiple Sclerosis Center of Excellence, Autoimmunity Center of Excellence, Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, MI, USA
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA
- Department of Physiology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
2
|
Kihara Y, Chun J. Molecular and neuroimmune pharmacology of S1P receptor modulators and other disease-modifying therapies for multiple sclerosis. Pharmacol Ther 2023; 246:108432. [PMID: 37149155 DOI: 10.1016/j.pharmthera.2023.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a neurological, immune-mediated demyelinating disease that affects people in the prime of life. Environmental, infectious, and genetic factors have been implicated in its etiology, although a definitive cause has yet to be determined. Nevertheless, multiple disease-modifying therapies (DMTs: including interferons, glatiramer acetate, fumarates, cladribine, teriflunomide, fingolimod, siponimod, ozanimod, ponesimod, and monoclonal antibodies targeting ITGA4, CD20, and CD52) have been developed and approved for the treatment of MS. All the DMTs approved to date target immunomodulation as their mechanism of action (MOA); however, the direct effects of some DMTs on the central nervous system (CNS), particularly sphingosine 1-phosphate (S1P) receptor (S1PR) modulators, implicate a parallel MOA that may also reduce neurodegenerative sequelae. This review summarizes the currently approved DMTs for the treatment of MS and provides details and recent advances in the molecular pharmacology, immunopharmacology, and neuropharmacology of S1PR modulators, with a special focus on the CNS-oriented, astrocyte-centric MOA of fingolimod.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, United States of America.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, United States of America
| |
Collapse
|
3
|
Reis-Mendes A, Carvalho F, Remião F, Sousa E, de Lourdes Bastos M, Costa VM. Autophagy (but not metabolism) is a key event in mitoxantrone-induced cytotoxicity in differentiated AC16 cardiac cells. Arch Toxicol 2023; 97:201-216. [PMID: 36216988 DOI: 10.1007/s00204-022-03363-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/11/2022] [Indexed: 01/19/2023]
Abstract
Mitoxantrone (MTX) is an antineoplastic agent used to treat advanced breast cancer, prostate cancer, acute leukemia, lymphoma and multiple sclerosis. Although it is known to cause cumulative dose-related cardiotoxicity, the underlying mechanisms are still poorly understood. This study aims to compare the cardiotoxicity of MTX and its' pharmacologically active metabolite naphthoquinoxaline (NAPHT) in an in vitro cardiac model, human-differentiated AC16 cells, and determine the role of metabolism in the cardiotoxic effects. Concentration-dependent cytotoxicity was observed after MTX exposure, affecting mitochondrial function and lysosome uptake. On the other hand, the metabolite NAPHT only caused concentration-dependent cytotoxicity in the MTT reduction assay. When assessing the effect of different inhibitors/inducers of metabolism, it was observed that metyrapone (a cytochrome P450 inhibitor) and phenobarbital (a cytochrome P450 inducer) slightly increased MTX cytotoxicity, while 1-aminobenzotriazole (a suicide cytochrome P450 inhibitor) decreased fairly the MTX-triggered cytotoxicity in differentiated AC16 cells. When focusing in autophagy, the mTOR inhibitor rapamycin and the autophagy inhibitor 3-methyladenine exacerbated the cytotoxicity caused by MTX and NAPHT, while the autophagy blocker, chloroquine, partially reduced the cytotoxicity of MTX. In addition, we observed a decrease in p62, beclin-1, and ATG5 levels and an increase in LC3-II levels in MTX-incubated cells. In conclusion, in our in vitro model, neither metabolism nor exogenously given NAPHT are major contributors to MTX toxicity as seen by the residual influence of metabolism modulators used on the observed cytotoxicity and by NAPHT's low cytotoxicity profile. Conversely, autophagy is involved in MTX-induced cytotoxicity and MTX seems to act as an autophagy inducer, possibly through p62/LC3-II involvement.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,Department of Biological Sciences, UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,Department of Biological Sciences, UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,Department of Biological Sciences, UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, 4450-208, Porto, Portugal
| | - Maria de Lourdes Bastos
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,Department of Biological Sciences, UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal. .,Department of Biological Sciences, UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
4
|
Mei KC, Liao YP, Jiang J, Chiang M, Khazaieli M, Liu X, Wang X, Liu Q, Chang CH, Zhang X, Li J, Ji Y, Melano B, Telesca D, Xia T, Meng H, Nel AE. Liposomal Delivery of Mitoxantrone and a Cholesteryl Indoximod Prodrug Provides Effective Chemo-immunotherapy in Multiple Solid Tumors. ACS NANO 2020; 14:13343-13366. [PMID: 32940463 PMCID: PMC8023019 DOI: 10.1021/acsnano.0c05194] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We developed a custom-designed liposome carrier for codelivery of a potent immunogenic cell death (ICD) stimulus plus an inhibitor of the indoleamine 2,3-dioxygenase (IDO-1) pathway to establish a chemo-immunotherapy approach for solid tumors in syngeneic mice. The carrier was constructed by remote import of the anthraquinone chemotherapeutic agent, mitoxantrone (MTO), into the liposomes, which were further endowed with a cholesterol-conjugated indoximod (IND) prodrug in the lipid bilayer. For proof-of-principle testing, we used IV injection of the MTO/IND liposome in a CT26 colon cancer model to demonstrate the generation of a robust immune response, characterized by the appearance of ICD markers (CRT and HMGB-1) as well as evidence of cytotoxic cancer cell death, mediated by perforin and granzyme B. Noteworthy, the cytotoxic effects involved natural killer (NK) cell, which suggests a different type of ICD response. The immunotherapy response was significantly augmented by codelivery of the IND prodrug, which induced additional CRT expression, reduced number of Foxp3+ Treg, and increased perforin release, in addition to extending animal survival beyond the effect of an MTO-only liposome. The outcome reflects the improved pharmacokinetics of MTO delivery to the cancer site by the carrier. In light of the success in the CT26 model, we also assessed the platform efficacy in further breast cancer (EMT6 and 4T1) and renal cancer (RENCA) models, which overexpress IDO-1. Encapsulated MTO delivery was highly effective for inducing chemo-immunotherapy responses, with NK participation, in all tumor models. Moreover, the growth inhibitory effect of MTO was enhanced by IND codelivery in EMT6 and 4T1 tumors. All considered, our data support the use of encapsulated MTO delivery for chemo-immunotherapy, with the possibility to boost the immune response by codelivery of an IDO-1 pathway inhibitor.
Collapse
Affiliation(s)
- Kuo-Ching Mei
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Michelle Chiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Mercedeh Khazaieli
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Liu
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Qi Liu
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Chong Hyun Chang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiao Zhang
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Juan Li
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Ying Ji
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Brenda Melano
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Donatello Telesca
- Department of Biostatistics, University of California, Los Angeles, California, 90095, United States
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Huan Meng
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, 90095, United States
| | - Andre E. Nel
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, 90095, United States
| |
Collapse
|
5
|
A Precise Nanostructure of Folate-Overhung Mitoxantrone DNA Tetrahedron for Targeted Capture Leukemia. NANOMATERIALS 2020; 10:nano10050951. [PMID: 32429472 PMCID: PMC7279534 DOI: 10.3390/nano10050951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/28/2020] [Accepted: 05/14/2020] [Indexed: 12/31/2022]
Abstract
Regular chemotherapy cannot eliminate leukemic cells, due to the sparse distribution of cancer cells in leukemia patients. Here, we report a precise nanostructure of folate-overhung mitoxantrone DNA tetrahedron that enables the treatment of leukemic cells by targeted action. Folate is used as a targeting molecule and synthesized with DNA strand in forming the folate-overhang DNA complement, and the complement is then separately base-paired onto six sides of the fabricated DNA tetrahedron. Mitoxantrone is used as an anticancer agent and intercalated into the double strands of the folate-overhung DNA tetrahedron for drug loading. The evaluation studies are performed on leukemia BALL-1 and K562 cells. The results demonstrate that the folate-overhung mitoxantrone DNA tetrahedra (approximately 25 nm) are able to target leukemic cells, transport across the nuclei membrane, induce the apoptosis, and enhance the overall efficacy of treating leukemic cells in vitro and in leukemia-bearing mice. This study provides a potential drug-containing DNA nanostructure, to clean the sparsely distributed leukemic cells in patients.
Collapse
|
6
|
Mao-Draayer Y, Thiel S, Mills EA, Chitnis T, Fabian M, Katz Sand I, Leite MI, Jarius S, Hellwig K. Neuromyelitis optica spectrum disorders and pregnancy: therapeutic considerations. Nat Rev Neurol 2020; 16:154-170. [PMID: 32080393 DOI: 10.1038/s41582-020-0313-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2020] [Indexed: 12/18/2022]
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are a type of neurological autoimmune disease characterized by attacks of CNS inflammation that are often severe and predominantly affect the spinal cord and optic nerve. The majority of individuals with NMOSD are women, many of whom are of childbearing age. Although NMOSD are rare, several small retrospective studies and case reports have indicated that pregnancy can worsen disease activity and might contribute to disease onset. NMOSD disease activity seems to negatively affect pregnancy outcomes. Moreover, some of the current NMOSD treatments are known to pose risks to the developing fetus and only limited safety data are available for others. Here, we review published studies regarding the relationship between pregnancy outcomes and NMOSD disease activity. We also assess the risks associated with using disease-modifying therapies for NMOSD during the course of pregnancy and breastfeeding. On the basis of the available evidence, we offer recommendations regarding the use of these therapies in the course of pregnancy planning in individuals with NMOSD.
Collapse
Affiliation(s)
- Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sandra Thiel
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tanuja Chitnis
- Department of Neurology, Brigham and Women's Hospital and Massachusetts General Hospital, Boston, MA, USA
| | - Michelle Fabian
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilana Katz Sand
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Isabel Leite
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Kerstin Hellwig
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
7
|
Oblad R, Doughty H, Lawson J, Christensen M, Kenealey J. Application of Mixture Design Response Surface Methodology for Combination Chemotherapy in PC-3 Human Prostate Cancer Cells. Mol Pharmacol 2018; 94:907-916. [PMID: 29884690 DOI: 10.1124/mol.117.111450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 06/07/2018] [Indexed: 12/17/2023] Open
Abstract
Combining chemotherapeutics to treat malignant tumors has been shown to be effective in preventing drug resistance, tumor recurrence, and reducing tumor size. We modeled combination drug therapy in PC-3 human prostate cancer cells using mixture design response surface methodology (MDRSM), a statistical technique designed to optimize compositions that we applied in a novel manner to design combinations of chemotherapeutics. Conventional chemotherapeutics (mitoxantrone, cabazitaxel, and docetaxel) and natural bioactive compounds (resveratrol, piperlongumine, and flavopiridol) were used in 12 different combinations containing three drugs at varying concentrations. Cell viability and cell cycle data were collected and used to plot response surfaces in MDRSM that identified the most effective concentrations of each drug in combination. MDRSM allows for extrapolation of data from three or more compounds in variable ratio combinations, unlike the Chou-Talalay method. MDRSM combinations were compared with combination index data from the Chou-Talalay method and were found to coincide. We propose MDRSM as an effective tool in devising combination treatments that can improve treatment effectiveness and increase treatment personalization, because MDRSM measures effectiveness rather than synergism, potentiation, or antagonism.
Collapse
Affiliation(s)
- Richard Oblad
- Department of Nutrition, Dietetics, and Food Science (R.O., H.D., M.C., J.K.) and Statistics (J.L.), Brigham Young University, Provo, Utah
| | - Hayden Doughty
- Department of Nutrition, Dietetics, and Food Science (R.O., H.D., M.C., J.K.) and Statistics (J.L.), Brigham Young University, Provo, Utah
| | - John Lawson
- Department of Nutrition, Dietetics, and Food Science (R.O., H.D., M.C., J.K.) and Statistics (J.L.), Brigham Young University, Provo, Utah
| | - Merrill Christensen
- Department of Nutrition, Dietetics, and Food Science (R.O., H.D., M.C., J.K.) and Statistics (J.L.), Brigham Young University, Provo, Utah
| | - Jason Kenealey
- Department of Nutrition, Dietetics, and Food Science (R.O., H.D., M.C., J.K.) and Statistics (J.L.), Brigham Young University, Provo, Utah
| |
Collapse
|
8
|
Emi TT, Barnes T, Orton E, Reisch A, Tolouei AE, Madani SZM, Kennedy SM. Pulsatile Chemotherapeutic Delivery Profiles Using Magnetically Responsive Hydrogels. ACS Biomater Sci Eng 2018; 4:2412-2423. [PMID: 30019005 PMCID: PMC6039960 DOI: 10.1021/acsbiomaterials.8b00348] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/15/2018] [Indexed: 12/17/2022]
Abstract
![]()
Pulsatile
chemotherapeutic delivery profiles may provide a number
advantages by maximizing the anticancer toxicity of chemotherapeutics,
reducing off-target side effects, and combating adaptive resistance.
While these temporally dynamic deliveries have shown some promise,
they have yet to be clinically deployed from implantable hydrogels,
whose localized deliveries could further enhance therapeutic outcomes.
Here, several pulsatile chemotherapeutic delivery profiles were tested
on melanoma cell survival in vitro and compared to constant (flatline)
delivery profiles of the same integrated dose. Results indicated that
pulsatile delivery profiles were more efficient at killing melanoma
cells than flatline deliveries. Furthermore, results suggested that
parameters like the duration of drug “on” periods (pulse
width), delivery rates during those periods (pulse heights), and the
number/frequency of pulses could be used to optimize delivery profiles.
Optimization of pulsatile profiles at tumor sites in vivo would require
hydrogel materials capable of producing a wide variety of pulsatile
profiles (e.g., of different pulse heights, pulse widths, and pulse
numbers). This work goes on to demonstrate that magnetically responsive,
biphasic ferrogels are capable of producing pulsatile mitoxantrone
delivery profiles similar to those tested in vitro. Pulse parameters
such as the timing and rate of delivery during “on”
periods could be remotely regulated through the use of simple, hand-held
magnets. The timing of pulses was controlled simply by deciding when
and for how long to magnetically stimulate. The rate of release during
pulse “on” periods was a function of the magnetic stimulation
frequency. These findings add to the growing evidence that pulsatile
chemotherapeutic delivery profiles may be therapeutically beneficial
and suggest that magnetically responsive hydrogels could provide useful
tools for optimizing and clinically deploying pulsatile chemotherapeutic
delivery profiles.
Collapse
Affiliation(s)
- Tania T Emi
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Tanner Barnes
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island 028881, United States
| | - Emma Orton
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island 028881, United States
| | - Anne Reisch
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island 028881, United States
| | - Anita E Tolouei
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - S Zahra M Madani
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Stephen M Kennedy
- Department of Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States.,Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island 028881, United States
| |
Collapse
|
9
|
Olusanya TOB, Haj Ahmad RR, Ibegbu DM, Smith JR, Elkordy AA. Liposomal Drug Delivery Systems and Anticancer Drugs. Molecules 2018; 23:E907. [PMID: 29662019 PMCID: PMC6017847 DOI: 10.3390/molecules23040907] [Citation(s) in RCA: 306] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/29/2018] [Accepted: 04/09/2018] [Indexed: 12/27/2022] Open
Abstract
Cancer is a life-threatening disease contributing to ~3.4 million deaths worldwide. There are various causes of cancer, such as smoking, being overweight or obese, intake of processed meat, radiation, family history, stress, environmental factors, and chance. The first-line treatment of cancer is the surgical removal of solid tumours, radiation therapy, and chemotherapy. The systemic administration of the free drug is considered to be the main clinical failure of chemotherapy in cancer treatment, as limited drug concentration reaches the tumour site. Most of the active pharmaceutical ingredients (APIs) used in chemotherapy are highly cytotoxic to both cancer and normal cells. Accordingly, targeting the tumour vasculatures is essential for tumour treatment. In this context, encapsulation of anti-cancer drugs within the liposomal system offers secure platforms for the targeted delivery of anti-cancer drugs for the treatment of cancer. This, in turn, can be helpful for reducing the cytotoxic side effects of anti-cancer drugs on normal cells. This short-review focuses on the use of liposomes in anti-cancer drug delivery.
Collapse
Affiliation(s)
- Temidayo O B Olusanya
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK.
| | - Rita Rushdi Haj Ahmad
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK.
| | - Daniel M Ibegbu
- Department of Medical Biochemistry, College of Medicine, University of Nigeria Enugu Campus, Nigeria.
| | - James R Smith
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| | - Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK.
| |
Collapse
|
10
|
Štenglová-Netíková IR, Petruželka L, Šťastný M, Štengl V. Anthracycline antibiotics derivate mitoxantrone-Destructive sorption and photocatalytic degradation. PLoS One 2018. [PMID: 29534071 PMCID: PMC5849306 DOI: 10.1371/journal.pone.0193116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nanostructured titanium(IV) oxide was used for the destructive adsorption and photocatalytic degradation of mitoxantrone (MTX), a cytostatic drug from the group of anthracycline antibiotics. During adsorption on a titania dioxide surface, four degradation products of MTX, mitoxantrone dicarboxylic acid, 1,4-dihydroxy-5-((2-((2-hydroxyethyl)amino)ethyl)amino)-8-((2-(methylamino)ethyl)amino)anthracene-9,10-dione, 1,4-dihydroxy-5,8-diiminoanthracene-9,10(5H,8H)-dione and 1,4-dihydroxy-5-imino-8-(methyleneamino)anthracene-9,10(5H,8H)-dione, were identified. In the case of photocatalytic degradation, only one degradation product after 15 min at m/z 472 was identified. This degradation product corresponded to mitoxantrone dicarboxylic acid, and complete mineralization was attained in one hour. Destructive adsorbent manganese(IV) oxide, MnO2, was used only for the destructive adsorption of MTX. Destructive adsorption occurred only for one degradation product, mitoxantrone dicarboxylic acid, against anatase TiO2.
Collapse
Affiliation(s)
| | - Luboš Petruželka
- Department of Oncology, 1 Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Martin Šťastný
- Department of Material Chemistry, Institute of Inorganic Chemistry ASCR, Husinec-Rez, Czech Republic
- Faculty of Environment, J.E.Purkyně University in Ústí nad Labem, Czech Republic
| | - Václav Štengl
- Department of Oncology, 1 Faculty of Medicine, Charles University in Prague, Czech Republic
- Department of Material Chemistry, Institute of Inorganic Chemistry ASCR, Husinec-Rez, Czech Republic
| |
Collapse
|
11
|
Teng Y, Wang Z, Li W, Yu J, Shan Z, Liang C, Wang S. Mitoxantrone suppresses vascular smooth muscle cell (VSMC) proliferation and balloon injury-induced neointima formation: An in vitro and in vivo study. Bosn J Basic Med Sci 2017; 17:339-348. [PMID: 28590233 DOI: 10.17305/bjbms.2017.2113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 11/16/2022] Open
Abstract
Neointima formation, which occurs after vascular injury due to vascular disease or interventions such as angioplasty and stent placement, is a complex process that involves multiple molecular and cellular mechanisms. The inhibition of neointima formation is vital to prevent restenosis of blood vessels. In the present study, we investigated whether the systemic administration of mitoxantrone can inhibit neointima formation, and evaluated the underlying mechanisms under in vitro and in vivo experimental conditions. In vitro, rat and human vascular smooth muscle cells (RVSMCs and HVSMCs) were stimulated with platelet-derived growth factor-BB (PDGF-BB) and treated with mitoxantrone or DMSO as a control. In vivo, 54 male Sprague-Dawley rats were subjected to carotid artery balloon injury and then intravenously administered with mitoxantrone. Cell proliferation was determined using the CCK-8 assay. Cell cycle analysis was performed using flow cytometry, and protein expression was analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis. We used monoclonal mouse anti-bromodeoxyuridine (BrdU) antibody for the detection of BrdU and anti-Topoisomerase II antibody for staining Type II topoisomerase (Topo II), one week after the ballon injury. In both RVSMCs and HVSMCs, mitoxantrone treatment induced Topo II degradation, as well as suppressed DNA replication, cell cycle progression, and VSMC proliferation. A reduction in intimal hyperplasia, intimal-to-medial area ratio, and Topo II level was observed in mitoxantrone-treated rats, as compared to the control (saline) group. Overall, our results indicate that systemic administration of mitoxantrone can reduce neointimal hyperplasia and, thus, represents a suitable option for restenosis treatment.
Collapse
Affiliation(s)
- Yuan Teng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | | | | | | | | | | | | |
Collapse
|
12
|
Hrynchak I, Sousa E, Pinto M, Costa VM. The importance of drug metabolites synthesis: the case-study of cardiotoxic anticancer drugs. Drug Metab Rev 2017; 49:158-196. [DOI: 10.1080/03602532.2017.1316285] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ivanna Hrynchak
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- CIIMAR – Centro Interdisciplinar de Investigação Marinha e Ambiental, Matosinhos, Portugal
| | - Madalena Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- CIIMAR – Centro Interdisciplinar de Investigação Marinha e Ambiental, Matosinhos, Portugal
| | - Vera Marisa Costa
- Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, UCIBIO, REQUIMTE (Rede de Química e Tecnologia), Universidade do Porto, Porto, Portugal
| |
Collapse
|
13
|
Thöne J, Thiel S, Gold R, Hellwig K. Treatment of multiple sclerosis during pregnancy - safety considerations. Expert Opin Drug Saf 2017; 16:523-534. [PMID: 28333552 DOI: 10.1080/14740338.2017.1311321] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Women with multiple sclerosis (MS) are treated early in the disease course with disease modifying therapies (DMT). Updated information is needed on pregnancy outcomes of DMT-exposed pregnancies and the effect of the drug withdrawal on MS disease activity. Areas covered: In this review, we will cover the most important updated management strategies in planning a pregnancy when having MS. Expert opinion: MS itself does not increase the risk of adverse pregnancy outcomes and does not negatively influence the long-term course of the disease. As MS became a treatable disease, management of DMTs before, during and after pregnancy is important. This requires updated knowledge on safety of DMTs as well as data of the effect on disease activity after drug withdrawal. A special challenge is the handling of women with highly active MS, as pregnancy might not be powerful enough to suppress the risk of rebound relapses. Exclusive breastfeeding is an option for many women who want to do so, but in cases of high disease activity and those women who do not want to breastfeed, early reintroduction of MS therapies should be considered.
Collapse
Affiliation(s)
- Jan Thöne
- a Department of Neurology , University Hospital Essen , Essen , Germany
| | - Sandra Thiel
- b Department of Neurology, St. Josef-Hospital , Ruhr-University Bochum , Bochum , Germany
| | - Ralf Gold
- b Department of Neurology, St. Josef-Hospital , Ruhr-University Bochum , Bochum , Germany
| | - Kerstin Hellwig
- b Department of Neurology, St. Josef-Hospital , Ruhr-University Bochum , Bochum , Germany
| |
Collapse
|
14
|
Reis-Mendes A, Gomes AS, Carvalho RA, Carvalho F, Remião F, Pinto M, Bastos ML, Sousa E, Costa VM. Naphthoquinoxaline metabolite of mitoxantrone is less cardiotoxic than the parent compound and it can be a more cardiosafe drug in anticancer therapy. Arch Toxicol 2016; 91:1871-1890. [PMID: 27629428 DOI: 10.1007/s00204-016-1839-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022]
Abstract
Mitoxantrone (MTX) is an antineoplastic agent used to treat several types of cancers and on multiple sclerosis, which shows a high incidence of cardiotoxicity. Still, the underlying mechanisms of MTX cardiotoxicity are poorly understood and the potential toxicity of its metabolites scarcely investigated. Therefore, this work aimed to synthesize the MTX-naphthoquinoxaline metabolite (NAPHT) and to compare its cytotoxicity to the parent compound in 7-day differentiated H9c2 cells using pharmacological relevant concentrations (0.01-5 µM). MTX was more toxic in equivalent concentrations in all cytotoxicity tests performed [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide reduction, neutral red uptake, and lactate dehydrogenase release assays] and times tested (24 and 48 h). Both MTX and NAPHT significantly decreased mitochondrial membrane potential in 7-day differentiated H9c2 cells after a 12-h incubation. However, energetic pathways were affected in a different manner after MTX or NAPHT incubation. ATP increased and lactate levels decreased after a 24-h incubation with MTX, whereas for the same incubation time and concentrations, NAPHT did not cause any significant effect. The increased activity of ATP synthase seems responsible for MTX-induced increases in ATP levels, as oligomycin (an inhibitor of ATP synthase) abrogated this effect on 5 µM MTX-incubated cells. 3-Methyladenine (an autophagy inhibitor) was the only molecule to give a partial protection against the cytotoxicity produced by MTX or NAPHT. To the best of our knowledge, this was the first broad study on NAPHT cardiotoxicity, and it revealed that the parent drug, MTX, caused a higher disruption in the energetic pathways in a cardiac model in vitro, whereas autophagy is involved in the toxicity of both compounds. In conclusion, NAPHT is claimed to largely contribute to MTX-anticancer properties; therefore, this metabolite should be regarded as a good option for a safer anticancer therapy since it is less cardiotoxic than MTX.
Collapse
Affiliation(s)
- A Reis-Mendes
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - A S Gomes
- UCIBIO-REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.,Lab. Química Orgânica e Farmacêutica, Dep. Química, Faculdade de Farmácia, U. Porto, Porto, Portugal
| | - R A Carvalho
- Centre for Functional Ecology, Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - F Carvalho
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - F Remião
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - M Pinto
- Lab. Química Orgânica e Farmacêutica, Dep. Química, Faculdade de Farmácia, U. Porto, Porto, Portugal.,CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - M L Bastos
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - E Sousa
- Lab. Química Orgânica e Farmacêutica, Dep. Química, Faculdade de Farmácia, U. Porto, Porto, Portugal.,CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - V M Costa
- UCIBIO-REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
15
|
Cicha I, Scheffler L, Ebenau A, Lyer S, Alexiou C, Goppelt-Struebe M. Mitoxantrone-loaded superparamagnetic iron oxide nanoparticles as drug carriers for cancer therapy: Uptake and toxicity in primary human tubular epithelial cells. Nanotoxicology 2015; 10:557-66. [DOI: 10.3109/17435390.2015.1095364] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
16
|
Evison BJ, Sleebs BE, Watson KG, Phillips DR, Cutts SM. Mitoxantrone, More than Just Another Topoisomerase II Poison. Med Res Rev 2015; 36:248-99. [PMID: 26286294 DOI: 10.1002/med.21364] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023]
Abstract
Mitoxantrone is a synthetic anthracenedione originally developed to improve the therapeutic profile of the anthracyclines and is commonly applied in the treatment of breast and prostate cancers, lymphomas, and leukemias. A comprehensive overview of the drug's molecular, biochemical, and cellular pharmacology is presented here, beginning with the cardiotoxic nature of its predecessor doxorubicin and how these properties shaped the pharmacology of mitoxantrone itself. Although mitoxantrone is firmly established as a DNA topoisomerase II poison within mammalian cells, it is now clear that the drug interacts with a much broader range of biological macromolecules both covalently and noncovalently. Here, we consider each of these interactions in the context of their wider biological relevance to cancer therapy and highlight how they may be exploited to further enhance the therapeutic value of mitoxantrone. In doing so, it is now clear that mitoxantrone is more than just another topoisomerase II poison.
Collapse
Affiliation(s)
- Benny J Evison
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Keith G Watson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Don R Phillips
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| |
Collapse
|
17
|
Shaul P, Steinbuch KB, Blacher E, Stein R, Fridman M. Exploring the Effects of Glycosylation and Etherification of the Side Chains of the Anticancer Drug Mitoxantrone. ChemMedChem 2015; 10:1528-38. [DOI: 10.1002/cmdc.201500274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 02/02/2023]
|
18
|
Mitochondrial cumulative damage induced by mitoxantrone: late onset cardiac energetic impairment. Cardiovasc Toxicol 2014; 14:30-40. [PMID: 24096626 DOI: 10.1007/s12012-013-9230-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitoxantrone (MTX) is a chemotherapeutic agent, which presents late irreversible cardiotoxicity. This work aims to highlight the mechanisms involved in the MTX-induced cardiotoxicity, namely the effects toward mitochondria using in vivo and in vitro studies. Male Wistar rats were treated with 3 cycles of 2.5 mg/kg MTX at day 0, 10, and 20. One treated group was euthanized on day 22 (MTX22) to evaluate the early MTX cardiac toxic effects, while the other was euthanized on day 48 (MTX48), to allow the evaluation of MTX late cardiac effects. Cardiac mitochondria isolated from 4 adult untreated rats were also used to evaluate in vitro the MTX (10 nM, 100 nM, and 1 μM) direct effects upon mitochondria functionality. Two rats of MTX48 died on day 35, and MTX treatment caused a reduction in relative body weight gain in both treated groups with no significant changes in water and food intake. Decreased levels of plasma total creatine kinase and CK-MB were detected in the MTX22 group, and increased plasma levels of lactate were seen in MTX48. Increased cardiac relative mass and microscopic changes were evident in both treated groups. Considering mitochondrial effects, for the first time, it was evidenced that MTX induced an increase in the complex IV and complex V activities in MTX22 group, while a decrease in the complex V activity was accompanied by the reduction in ATP content in the MTX48 rats. No alterations in mitochondria transmembrane potential were found in isolated mitochondria from MTX48 rats or in isolated mitochondria directly incubated with MTX. This study highlights the relevance of the cumulative MTX-induced in vivo mitochondriopathy to the MTX cardiotoxicity.
Collapse
|
19
|
Shaaban S, Negm A, Ibrahim EE, Elrazak AA. Chemotherapeutic agents for the treatment of hepatocellular carcinoma: efficacy and mode of action. Oncol Rev 2014; 8:246. [PMID: 25992234 PMCID: PMC4419609 DOI: 10.4081/oncol.2014.246] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/06/2014] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a dreaded malignancy that every year causes half a million deaths worldwide. Being an aggressive cancer, its incidence exceeds 700,000 new cases per year worldwide with a median survival of 6-8 months. Despite advances in prognosis and early detection, effective HCC chemoprevention or treatment strategies are still lacking, therefore its dismal survival rate remains largely unchanged. This review will characterize currently available chemotherapeutic drugs used in the treatment of HCC. The respective mode(s) of action, side effects and recommendations will be also described for each drug.
Collapse
Affiliation(s)
- Saad Shaaban
- Department of Chemistry, Mansoura University , Egypt
| | - Amr Negm
- Department of Biochemistry, Mansoura University , Egypt
| | | | | |
Collapse
|
20
|
Neuhaus O, Kieseier BC, Hartung HP. Mitoxantrone (Novantrone®) in multiple sclerosis: new insights. Expert Rev Neurother 2014; 4:17-26. [PMID: 15853611 DOI: 10.1586/14737175.4.1.17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The conclusions of a recent study of mitoxantrone (Novantrone) in multiple sclerosis and the approval of several health authorities support its use in patients with active relapsing-remitting or secondary progressive multiple sclerosis. This drug profile provides an outline on relevant preclinical and clinical studies, discusses relevant side effects of the compound and places mitoxantrone in the context of other therapeutic approaches available against this disabling disorder.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Heinrich Heine University, Moorenstrasse 5, D 40225 Düsseldorf, Germany.
| | | | | |
Collapse
|
21
|
Rossato LG, Costa VM, Dallegrave E, Arbo M, Dinis-Oliveira RJ, Santos-Silva A, Duarte JA, de Lourdes Bastos M, Palmeira C, Remião F. Cumulative mitoxantrone-induced haematological and hepatic adverse effects in a subchronic in vivo study. Basic Clin Pharmacol Toxicol 2013; 114:254-62. [PMID: 24119282 DOI: 10.1111/bcpt.12143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 09/10/2013] [Indexed: 01/16/2023]
Abstract
Mitoxantrone (MTX) is an antineoplastic agent that can induce hepato- and haematotoxicity. This work aimed to investigate the occurrence of cumulative early and late MTX-induced hepatic and haematological disturbances in an vivo model. A control group and two groups treated with three cycles of 2.5 mg/kg MTX at days 0, 10 and 20 were formed. One of the treated groups suffered euthanasia on day 22 (MTX22) to evaluate early MTX toxic effects, while the other suffered euthanasia on day 48 (MTX48), to allow the evaluation of MTX late effects. An early immunosuppression with a drop in the IgG levels was observed, causing a slight decrease in the plasma total protein content. The early bone marrow depression was followed by signs of recovery in MTX48. The genotoxic potential of MTX was demonstrated by the presence of several micronuclei in MTX22 leucocytes. Increases in plasma iron and cholesterol levels in the MTX22 rats were observed, while in both groups increases in the unconjugated bilirubin, C4 complement, and decreases in the triglycerides, alanine aminotransferase, alkaline phosphatase and transferrin were found in plasma samples. On MTX 48, the liver histology showed more hepatotoxic signs, the hepatic levels of reduced and oxidized glutathione were increased, and ATP hepatic levels were decreased. However, the hepatic total protein levels were decreased only in the livers of MTX22 group. Results demonstrated the MTX genotoxic effects, haemato- and direct hepatotoxicity. While the haematological toxicity is ameliorated with time, the same was not observed in the hepatic injury.
Collapse
Affiliation(s)
- Luciana G Rossato
- REQUIMTE, Toxicology Laboratory, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Costa VM, Carvalho F, Duarte JA, Bastos MDL, Remião F. The Heart As a Target for Xenobiotic Toxicity: The Cardiac Susceptibility to Oxidative Stress. Chem Res Toxicol 2013; 26:1285-311. [PMID: 23902227 DOI: 10.1021/tx400130v] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Vera Marisa Costa
- REQUIMTE (Rede de Química e Tecnologia),
Laboratório de Toxicologia, Departamento de Ciências
Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia),
Laboratório de Toxicologia, Departamento de Ciências
Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | | | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia),
Laboratório de Toxicologia, Departamento de Ciências
Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Fernando Remião
- REQUIMTE (Rede de Química e Tecnologia),
Laboratório de Toxicologia, Departamento de Ciências
Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
23
|
Rossato LG, Costa VM, de Pinho PG, Arbo MD, de Freitas V, Vilain L, de Lourdes Bastos M, Palmeira C, Remião F. The metabolic profile of mitoxantrone and its relation with mitoxantrone-induced cardiotoxicity. Arch Toxicol 2013; 87:1809-20. [DOI: 10.1007/s00204-013-1040-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 03/08/2013] [Indexed: 01/26/2023]
|
24
|
An G, Morris ME. A physiologically based pharmacokinetic model of mitoxantrone in mice and scale-up to humans: a semi-mechanistic model incorporating DNA and protein binding. AAPS JOURNAL 2012; 14:352-64. [PMID: 22451016 DOI: 10.1208/s12248-012-9344-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 03/01/2012] [Indexed: 11/30/2022]
Abstract
We conducted a pharmacokinetic (PK) study of mitoxantrone (Novantrone®), a clinically well-established anticancer agent, in mice and developed a mechanism-based PBPK (physiologically based pharmacokinetic) model to describe its disposition. Mitoxantrone concentrations in plasma and six organs (lung, heart, liver, kidney, spleen, and brain) were determined after a 5 mg/kg i.v. dose. We evaluated three different PBPK models in order to characterize our experimental data: model 1 containing Kp values, model 2 incorporating a deep binding compartment, and model 3 incorporating binding of mitoxantrone to DNA and protein. Among the three models, only model 3 with DNA and protein binding captured all the experimental data well. The estimated binding affinity for DNA (K (DNA)) and protein (K (macro)) were 0.0013 and 1.44 μM, respectively. Predicted plasma and tissue AUC values differed from observed values by <19 %, except for heart (60 %). Model 3 was further used to simulate plasma mitoxantrone concentrations in humans for a 12-mg/m(2) dose, using human physiological parameters. The simulated results generally agreed with the observed time course of mitoxantrone plasma concentrations in patients after a standard dose of 12 mg/m(2). In summary, we reported for the first time a mechanism-based PBPK model of mitoxantrone incorporating macromolecule binding which may have clinical applicability in optimizing clinical therapy. Since mitoxantrone is a substrate of the efflux transporters ABCG2 and ABCB1, the incorporation of efflux transporters may also be necessary to characterize the data obtained in low-dose studies.
Collapse
Affiliation(s)
- Guohua An
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, 517 Hochstetter Hall, Amherst, New York 14260-1200, USA
| | | |
Collapse
|
25
|
Collongues N, de Seze J. Current and future treatment approaches for neuromyelitis optica. Ther Adv Neurol Disord 2011; 4:111-21. [PMID: 21694808 DOI: 10.1177/1756285611398939] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neuromyelitis optica (NMO) is an inflammatory disease of the central nervous system (CNS) characterized by severe attacks of optic neuritis and myelitis, and which, unlike multiple sclerosis (MS), commonly spares the brain in the early stages. NMO used to be considered as a special form of MS. During the past 10 years, however, the two diseases have been shown to be clearly different. NMO is a B-cell-mediated disease associated with anti-aquaporin-4 antibodies in many cases and its pathophysiology seems to be near the acute lesion of necrotizing vasculitis. Assessment of prevalence shows that NMO is far less frequent than MS, which explains the absence of randomized clinical trials and NMO treatment strategies validated by evidence-based medicine. Recently, many data have been published that suggest that the therapeutic option in NMO should be immunosuppressive rather than immunomodulatory drugs. In the present study, after a brief overview of NMO, we review therapeutic studies and propose new therapeutic strategies in the relapse and disease-modifying fields.
Collapse
Affiliation(s)
- Nicolas Collongues
- Centre d'investigation Clinique, INSERM 002, Nouvel hopital civil, 1 place de l'hopital, BP 426, 67091 Strasbourg cedex, France; Department of Neurology, University Hospitals of Strasbourg, Strasbourg, France
| | | |
Collapse
|
26
|
5,7-Dimethoxyflavone and multiple flavonoids in combination alter the ABCG2-mediated tissue distribution of mitoxantrone in mice. Pharm Res 2011; 28:1090-9. [PMID: 21279423 DOI: 10.1007/s11095-011-0368-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Accepted: 01/10/2011] [Indexed: 10/18/2022]
Abstract
PURPOSE The objective of our study was to investigate the effect of 5,7-DMF on the accumulation of mitoxantrone (MX) in BCRP-expressing normal cells and to investigate its impact on the PK and tissue distribution of MX in mice. METHODS The in vitro effect of 5,7-DMF on MX accumulation was examined in MDCK cells transfected with BCRP. The pharmacokinetic and tissue distribution of mitoxantrone, with and without co-administration of 5,7-DMF or multiple flavonoid combinations, were determined in mice. RESULTS In the presence of 2.5 μM or 25 μM of 5,7-DMF, the intracellular concentration of MX was significantly increased in MDCK/Bcrp1 and MDCK/BCRP cells, but not in MDCK/Mock cells. The AUC values of MX in several tissues were significantly increased when MX was co-administered with 5,7-DMF. The most substantial elevations of MX AUC in the presence of 5,7-DMF occurred in the liver (94.5%) and kidneys (61.9%), which is in apparent agreement with the relatively high levels of mouse Bcrp1 expression in these two tissues. CONCLUSIONS Bcrp1-mediated DMF-MX interactions occur both in vitro and in vivo. 5,7-DMF represents a novel and very promising chemosensitizing agent for the BCRP-mediated MDR due to its low toxicity and potent BCRP inhibition.
Collapse
|
27
|
An G, Morris ME. Effects of the isoflavonoid biochanin A on the transport of mitoxantrone in vitro and in vivo. Biopharm Drug Dispos 2010; 31:340-50. [PMID: 20535833 DOI: 10.1002/bdd.717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The aim of our study was to investigate the effect of biochanin A on the accumulation and transport of mitoxantrone in breast cancer resistance protein (BCRP)-expressing normal cells and its impact on the pharmacokinetics (PK) and tissue distribution of mitoxantrone. In accumulation studies, the intracellular level of mitoxantrone was significantly increased in the presence of 2.5 or 25 microM of biochanin A in both murine and human BCRP-expressing Madin-Darby canine kidney (MDCK) cells, with no effect in corresponding MDCK/Mock cells. In bi-directional transport studies, the P(app,B-A) value of mitoxantrone with biochanin A co-treatment was much lower (6.66+/-0.84x10(-7) cm/s) than that in the absence of biochanin A (21.4+/-4.14x10(-7) cm/s), indicating inhibition of Bcrp1-mediated efflux. To evaluate whether our in vitro results might translate into an in vivo interaction, mitoxantrone PK and tissue distribution, with and without co-administration of biochanin A, was investigated. In contrast to our in vitro results, biochanin A (10 mg/kg, i.v.) had no impact on the concentration of mitoxantrone in plasma and most tissues collected (brain, heart, liver and lung). Surprisingly, the concentrations of mitoxantrone in spleen and kidney were even decreased when biochanin A was co-administered. Interestingly, it was found that the intracellular fluorescence of mitoxantrone was decreased 31.9% when co-incubated with 10 microM biochanin A in P-glycoprotein (P-gp) expressing MCF-7/ADR cells, indicating potential P-gp stimulation. The species difference of the inhibitory effect of biochanin A on BCRP, the extensive metabolism of biochanin A, as well as the stimulation effect of biochanin A on P-gp, may contribute to this in vitro-in vivo disconnect.
Collapse
Affiliation(s)
- Guohua An
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, 14260, USA
| | | |
Collapse
|
28
|
Zhang LK, Hou SX, Zhang JQ, Hu WJ, Wang CY. Preparation, characterization, and in vivo evaluation of mitoxantrone-loaded, folate-conjugated albumin nanoparticles. Arch Pharm Res 2010; 33:1193-8. [PMID: 20803122 DOI: 10.1007/s12272-010-0809-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 02/22/2010] [Accepted: 04/11/2010] [Indexed: 10/19/2022]
Abstract
Folic acid was covalently conjugated to bovine serum albumin nanoparticles (BSANP) to target the nanoparticles to SKOV3 cells expressing folate receptors. Mitoxantrone was incorporated into the folate-conjugated albumin nanoparticles, and the final nanoparticle size was 68 nm, as measured by a laser light scattering particle analyzer. The cytotoxic activity of mitoxantrone- loaded, folate-conjugated albumin nanoparticles (MTO-BSANP-folate), which was quantitated by (3)H-thymidine incorporation, was higher than mitoxantrone-loaded BSANP (MTO-BSANP) and MTO solution, and could be inhibited by free folic acid. MTO-BSANPfolate may be endocytosed via the folate receptor on the surface of SKOV3 cells. MTO-BSANPfolate also inhibited tumor growth better than the MTO-BSANP and MTO solution in vivo. These results indicate that folate-conjugated BSANP may have therapeutic potential as a vector for anticancer drugs in cancer chemotherapy.
Collapse
Affiliation(s)
- Liang-ke Zhang
- Institute of Life Sciences, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.
| | | | | | | | | |
Collapse
|
29
|
Schedule treatment design and quantitative in vitro evaluation of chemotherapeutic combinations for metastatic prostate cancer therapy. Cancer Chemother Pharmacol 2010; 67:275-84. [DOI: 10.1007/s00280-010-1315-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 03/23/2010] [Indexed: 10/19/2022]
|
30
|
Effects of single and multiple flavonoids on BCRP-mediated accumulation, cytotoxicity and transport of mitoxantrone in vitro. Pharm Res 2010; 27:1296-308. [PMID: 20369276 DOI: 10.1007/s11095-010-0108-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 03/01/2010] [Indexed: 02/06/2023]
Abstract
PURPOSE The objective of our study was to investigate the effect of single and multiple flavonoids on the accumulation and cytotoxicity of mitoxantrone in BCRP-overexpressing breast cancer cells and on the transport of mitoxantrone in BCRP-expressing normal cells. METHODS The effect of flavonoids on mitoxantrone accumulation and cytotoxicity was studied in the human breast cancer MCF-7 MX100 cell line. Mitoxantrone transport in the presence of flavonoids was studied in human and murine BCRP-transfected MDCK cell lines, and mitoxantrone concentrations were determined by HPLC. RESULTS Our results demonstrated that multiple flavonoid combinations act additively and exhibit strong BCRP inhibition for increasing mitoxantrone accumulation in breast cancer cells. Kaempferide, biochanin A, 5,7-dimethoxyflavone, and 8-methylflavone greatly increased the cytotoxicity of mitoxantrone in BCRP-overexpressing breast cancer cells. Additionally, the basolateral-to-apical membrane-directed transport of mitoxantrone in murine Bcrp1- and human BCRP-expressing MDCK cells, in the presence of 2.5 microM of these flavonoids, was also significantly decreased. CONCLUSION The results indicate that flavonoids are potent BCRP inhibitors and that they exert additive effects when used in combination. Flavonoids demonstrate MDR-reversing effects, but also may influence the disposition of mitoxantrone and cause pharmacokinetic interactions.
Collapse
|
31
|
Gruber A, Liliemark J, Tidefelt U, Knochenhauer E, Arestrom I, Sundman-Engberg B, Bjorkholm M, Paul C, Peterson C. Pharmacokinetics of Mitoxantrone in Plasma and Leukemic Cells During Treatment of Patients with Acute Non-Lymphocytic Leukemia. Leuk Lymphoma 2009. [DOI: 10.3109/10428199209053588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Astrid Gruber
- Division of Hematology and Immunology, Department of Medicine, Stockholm, Sweden
- Department of Clinical Pharmacology, Karolinska Hospital, Stockholm, Sweden
| | - Jan Liliemark
- Department of Clinical Pharmacology, Karolinska Hospital, Stockholm, Sweden
- Division of Clinical Hematology and Oncology, Department of Medicine, Huddinge Hospital, Huddinge, Sweden
| | - Ulf Tidefelt
- Division of Clinical Hematology and Oncology, Department of Medicine, Huddinge Hospital, Huddinge, Sweden
| | - Eva Knochenhauer
- Department of Clinical Pharmacology, Karolinska Hospital, Stockholm, Sweden
| | - Irene Arestrom
- Department of Clinical Pharmacology, Karolinska Hospital, Stockholm, Sweden
| | - Britt Sundman-Engberg
- Division of Clinical Hematology and Oncology, Department of Medicine, Huddinge Hospital, Huddinge, Sweden
| | - Magnus Bjorkholm
- Division of Hematology and Immunology, Department of Medicine, Stockholm, Sweden
| | - Christer Paul
- Division of Clinical Hematology and Oncology, Department of Medicine, Huddinge Hospital, Huddinge, Sweden
| | - Curt Peterson
- Department of Clinical Pharmacology, Karolinska Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Trédan O, Garbens AB, Lalani AS, Tannock IF. The hypoxia-activated ProDrug AQ4N penetrates deeply in tumor tissues and complements the limited distribution of mitoxantrone. Cancer Res 2009; 69:940-7. [PMID: 19176397 DOI: 10.1158/0008-5472.can-08-0676] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypoxic tumor cells are likely to be resistant to conventional chemotherapy, in large part because many anticancer drugs are unable to penetrate into poorly oxygenated tumor tissue. Here, we used quantitative immunofluorescence to study the distribution of mitoxantrone and AQ4N in tumor tissue. AQ4N is a prodrug activated under hypoxic conditions to AQ4, which is structurally similar to mitoxantrone and inhibits topoisomerase II. We characterized the penetration of mitoxantrone and AQ4N/AQ4 through multilayered cell cultures (MCC) and in relation to blood vessels and hypoxic regions in human tumor xenografts. We also studied tumor growth delay after treatment with each agent alone and with the combination. In both MCC and xenografts, mitoxantrone is taken up by proximal cells and penetrates slowly to distant regions. In contrast, AQ4N rapidly penetrates MCC and tumor tissue in vivo, and AQ4N (or its reduced form AQ4) is detected at high concentration within hypoxic regions. The targeting of mitoxantrone to oxygenated regions and AQ4N/AQ4 to hypoxic tumor regions results in effective drug exposure over the entire tumor after combined treatment and increases tumor growth delay compared with either drug alone. The combination of a clinically used anticancer drug with limited tissue penetration and a structurally related drug activated in regions of tumor hypoxia is an effective strategy to overcome chemoresistance due to the tumor microenvironment. This study supports clinical evaluation of AQ4N in combination with conventional anticancer agents, such as mitoxantrone.
Collapse
Affiliation(s)
- Olivier Trédan
- Division of Medical Oncology and Hematology, Princess Margaret Hospital and University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
33
|
Bally MB, Lim H, Cullis PR, Mayer LD. Controlling the Drug Delivery Attributes of Lipid-Based Drug Formulations. J Liposome Res 2008. [DOI: 10.3109/08982109809035537] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Schwendener R, Horber D, Rentsch K, Hänseler E, Pestalozzi B, Sauter C. Preclinical and Clinical Experience with Liposome-Encapsulated Mitoxantrone. J Liposome Res 2008. [DOI: 10.3109/08982109409037063] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Boster A, Edan G, Frohman E, Javed A, Stuve O, Tselis A, Weiner H, Weinstock-Guttman B, Khan O. Intense immunosuppression in patients with rapidly worsening multiple sclerosis: treatment guidelines for the clinician. Lancet Neurol 2008; 7:173-83. [PMID: 18207115 DOI: 10.1016/s1474-4422(08)70020-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Several lines of evidence link immunosuppression to inflammation in patients with multiple sclerosis (MS) and provide a rationale for the increasing use of immunosuppressive drugs in the treatment of MS. Treatment-refractory, clinically active MS can quickly lead to devastating and irreversible neurological disability and treating these patients can be a formidable challenge to the clinician. Patients with refractory MS have been treated with intense immunosuppression, such as cyclophosphamide or mitoxantrone, or with autologous haematopoeitic stem cell transplants. Evidence shows that intense immunosuppression might be effective in patients who are unresponsive to immunomodulating therapy, such as interferon beta and glatiramer acetate. Natalizumab, a new addition to the armamentarium for treating MS, might also have a role in the treatment of this MS phenotype. This Review describes the use of intense immunosuppressant drugs and natalizumab in patients with rapidly worsening MS and provides clinicians with guidelines for the use of these drugs in this patient group.
Collapse
Affiliation(s)
- Aaron Boster
- The Multiple Sclerosis Clinical Research Center, Department of Neurology, Wayne State University School of Medicine, and The Detroit Medical Center, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Severe bladder contracture leading to cystectomy after intravesical mitoxantrone chemotherapy. Anticancer Drugs 2008; 19:325-8. [DOI: 10.1097/cad.0b013e3282f421fe] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
38
|
Neuhaus O, Kieseier BC, Hartung HP. Pharmacokinetics and pharmacodynamics of the interferon-betas, glatiramer acetate, and mitoxantrone in multiple sclerosis. J Neurol Sci 2007; 259:27-37. [PMID: 17391705 DOI: 10.1016/j.jns.2006.05.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/25/2006] [Accepted: 05/01/2006] [Indexed: 11/15/2022]
Abstract
Five disease-modifying agents are currently approved for long-term treatment of multiple sclerosis (MS), namely three interferon-beta preparations, glatiramer acetate, and mitoxantrone(1). Pharmacokinetics describes the fate of drugs in the human body by studying their absorption, distribution, metabolism and excretion. Pharmacodynamics is dedicated to the mechanisms of action of drugs. The understanding of the pharmacokinetics and pharmacodynamics of the approved disease-modifying agents against MS is of importance as it might contribute to the development of future derivatives with a potentially higher efficacy and a more favourable safety profile. This article reviews data thus far present both on the pharmacokinetics as well as on the putative mechanisms of action of the interferon-betas, glatiramer acetate, and mitoxantrone in the immunopathogenesis of MS.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany.
| | | | | |
Collapse
|
39
|
Lim SJ, Cormier JN, Feig BW, Mansfield PF, Benjamin RS, Griffin JR, Chase JL, Pisters PWT, Pollock RE, Hunt KK. Toxicity and Outcomes Associated with Surgical Cytoreduction and Hyperthermic Intraperitoneal Chemotherapy (HIPEC) for Patients with Sarcomatosis. Ann Surg Oncol 2007; 14:2309-18. [PMID: 17541691 DOI: 10.1245/s10434-007-9463-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 04/24/2007] [Indexed: 01/28/2023]
Abstract
BACKGROUND Treatment of peritoneal recurrence following surgical resection of intra-abdominal sarcomas presents a significant challenge to clinicians. Historically, treatment with systemic chemotherapy has been ineffective and surgical resection alone has not been durable. We prospectively evaluated the feasibility of cytoreductive surgery followed by hyperthermic intraperitoneal chemotherapy (HIPEC) with cisplatin (CDDP) alone or in combination with mitoxantrone (MITOX) for the treatment of sarcomatosis. METHODS Two phase I trials of HIPEC were conducted (1998-2003). Eligible patients with evidence of sarcomatosis underwent cytoreductive surgery followed by HIPEC. In the first trial, CDDP dosing was established as 90 mg/m2 with a perfusate time of 90 minutes and temperature of 41 degrees C. In the second trial, MITOX (20 mg/m2) was instilled following perfusion with CDDP. Toxicity, efficacy, and quality of life (QOL) were evaluated. RESULTS A total of 28 patients were enrolled in the two trials. We noted a higher overall toxicity score and complication rate with combination CDDP/MITOX versus CDDP alone and shorter overall survival duration (5.5 months vs 16.9 months, respectively). In addition, local recurrence rates were similar in both groups (CDDP 79% vs CDDP/MITOX 68%). As expected, QOL scores at 6-8 weeks following HIPEC were 15-25% lower than the baseline scores; however, they returned to baseline at 3-6 months. CONCLUSIONS Although the HIPEC technique is feasible for patients with sarcomatosis, it is associated with significant toxicity and limited clinical benefit. Combination CDDP/MITOX failed to demonstrate any benefit over CDDP alone; moreover, there was an increase in toxicity.
Collapse
Affiliation(s)
- Sherry J Lim
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Niang M, Melka M, Stoklasová A, Cerman J, Tomsík P. Evaluation of the antineoplastic activity of mitoxantrone–l-carnitine combination therapy on an experimental solid form of ehrlich tumour in mice. Pharmacol Res 2006; 54:447-51. [PMID: 17049876 DOI: 10.1016/j.phrs.2006.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 08/13/2006] [Accepted: 09/12/2006] [Indexed: 11/20/2022]
Abstract
We have commenced a series of experiments to evaluate the effect of carnitine derivatives on the antineoplastic activity of mitoxantrone (MX) on various animal cancers. This report describes the therapeutic effect of MX in combination with l-carnitine (LCAR) on the growth of a solid form of Ehrlich tumour inoculated into mice. LCAR was administered subcutaneously at doses of either 200 or 100mgkg(-1) on day 6 and 13 after tumour inoculation, 1h prior to the treatment with MX. Mitoxantrone was administered intravenously at doses of 3 or 6mgkg(-1). We found that LCAR had no potentiating effect on the efficacy of MX, in terms of either slowing tumour growth or increasing the survival of mice. Nevertheless, therapeutic effects can be assumed at higher doses of both drugs based on values calculated from an index of relative hazards.
Collapse
Affiliation(s)
- M Niang
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Králové, Charles University in Prague, Simkova 870, 500 38 Hradec Králové, Czech Republic.
| | | | | | | | | |
Collapse
|
41
|
|
42
|
Neuhaus O, Kieseier BC, Hartung HP. Therapeutic role of mitoxantrone in multiple sclerosis. Pharmacol Ther 2005; 109:198-209. [PMID: 16095713 DOI: 10.1016/j.pharmthera.2005.07.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 07/07/2005] [Indexed: 11/26/2022]
Abstract
Mitoxantrone is approved by several health authorities for treatment of active forms of relapsing-remitting or secondary progressive multiple sclerosis (SPMS). This review provides an outline on relevant preclinical as well as clinical studies, places mitoxantrone in the context of other therapeutic approaches against multiple sclerosis (MS), and discusses relevant side effects. The current knowledge of the putative mechanisms of action of the compound is discussed.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Heinrich Heine University, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| | | | | |
Collapse
|
43
|
Dirsch O, Dahmen U, Gu YL, Ji Y, Karoussos IA, Wieneke H, Erbel R. Preliminary investigation of mitoxantrone coating on stent-grafts to inhibit neointimal proliferation. J Endovasc Ther 2005; 12:479-85. [PMID: 16048380 DOI: 10.1583/04-1444mr.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE To investigate the inhibition of neointimal proliferation induced by a stent-graft loaded with mitoxantrone. METHODS Stent-grafts with and without mitoxantrone loading (150 microg per device) were inserted into the carotid artery of 7 and 6 rabbits, respectively. After an observation period of 28 days, the animals were sacrificed, and a detailed morphological and morphometric workup of the stented vessels was performed. RESULTS Uncoated stent-grafts induced a thick neointima (median diameter 97+/-99 microm), whereas no neointima formed in mitoxantrone-loaded stent-grafts. However, the loaded devices were not fully covered by an endothelial layer. The underlying media was significantly thinner (31.8+/-5.6 versus 48.6+/-3.3 microm, p=0.002) and showed a widespread loss of smooth muscle cells. CONCLUSIONS Mitoxantrone loading of a stent-graft inhibited the formation of a neointima. However, important regenerative processes were prevented as well, indicating a local overdose. More experiments using lower doses are warranted.
Collapse
Affiliation(s)
- Olaf Dirsch
- Institute of Pathology, University of Cologne, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Ugwu S, Zhang A, Parmar M, Miller B, Sardone T, Peikov V, Ahmad I. Preparation, characterization, and stability of liposome-based formulations of mitoxantrone. Drug Dev Ind Pharm 2005; 31:223-9. [PMID: 15773289 DOI: 10.1081/ddc-200047850] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The preparation, characterization, and stability of lyophilized liposome-based formulation of mitoxantrone was investigated. Mitoxantrone was entrapped inside small, unilamellar liposomes composed of dioleoylphosphocholine (DOPC), cholesterol, and cardiolipin. The mean vesicle size and drug entrapment efficiency of the liposomes were approximately 150 nm and approximately 99%, respectively. Less than 1% of drug was lost and mean vesicle size remained unchanged after sterile filtration. The pre-lyophilized (pre-lyo) formulations were characterized by a differential scanning calorimetric (DSC) method. Results showed that the glass transition temperatures (Tg) increased as the molar ratios of sucrose:lipid and trehalose:lipid in the formulations were increased. The maximum Tg' of the pre-lyo formulations containing 10:1 sucrose:lipid and trehalose:lipid molar ratios were -37C and -41C, respectively. After reconstitution of the lyophilized cake of the sucrose-containing formulation, the mean vesicle size was comparable to pre-lyo liposome size. In vitro release studies showed that less than 2% of mitoxantrone was released after an extensive dialysis against phosphate buffered saline (PBS) at 37C, indicating that the mitoxantrone was highly associated and retained inside the liposomes. Short-term stability studies of the sucrose-containing formulations revealed that the reconstituted and eight-fold diluted formulations were stable for up to 8 hours at room temperature. Long-term stability studies of lyophilized liposomal mitoxantrone showed that the lyophilized formulation was stable for up to 13 months after storage at refrigerated condition.
Collapse
|
45
|
Bozdağ S, Çapan Y, Vural I, Dalkara T, Dogan A, Guc D, Hincal A, DeLuca P. Formulation and in vitro bioactivity of mitoxantrone-loaded biodegradable microspheres on rat glioma (RG2) cells. J Drug Deliv Sci Technol 2005. [DOI: 10.1016/s1773-2247(05)50032-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
46
|
Martincic D, Hande KR. Topoisomerase II inhibitors. CANCER CHEMOTHERAPY AND BIOLOGICAL RESPONSE MODIFIERS 2005; 22:101-21. [PMID: 16110609 DOI: 10.1016/s0921-4410(04)22005-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Danko Martincic
- Vanderbilt/Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
47
|
Abstract
Mitoxantrone (Novantrone), a synthetic anthracenedione derivative, is an antineoplastic, immunomodulatory agent. Its presumed mechanism of action in patients with multiple sclerosis (MS) is via immunomodulatory mechanisms, although these remain to be fully elucidated. Intravenous mitoxantrone treatment improved neurological disability and delayed progression of MS in patients with worsening relapsing-remitting (RR) [also termed progressive-relapsing (PR) MS] or secondary-progressive (SP) disease. In a pivotal randomised, double-blind, multicentre trial, mitoxantrone 12 mg/m(2) administered once every 3 months for 2 years provided significant improvements in neurological disability ratings, including Kurtzke Expanded Disability Status Scale (EDSS), Ambulatory Index (AI) and Standardised Neurological Status (SNS) scores, compared with placebo. The drug also significantly reduced the mean number of corticosteroid-treated relapses and prolonged the time to the first treated relapse, with the beneficial effects on disease progression supported by magnetic resonance imaging. Post hoc analyses suggest that the benefits associated with mitoxantrone treatment may be sustained for at least 12 months after cessation of treatment, mean changes from baseline at 36 months in EDSS, AI and SNS scores of 0.10, 0.61 and 0.19, respectively, in the mitoxantrone group versus 0.46, 1.13 and 3.38 with placebo. Concomitant intravenous mitoxantrone 20mg plus intravenous methylprednisolone 1g once every month for 6 months was more effective than intravenous methylprednisolone monotherapy in preventing the development of new gadolinium-enhanced lesions in patients with very active RRMS or SPMS. The drug was generally well tolerated in patients with MS. Adverse events were generally mild to moderate in severity and usually resolved upon discontinuation of treatment or with appropriate pharmacotherapy. At the recommended dosage, mitoxantrone appears to have a low potential to cause cardiotoxicity. In conclusion, intravenous mitoxantrone reduces the relapse rate and slows progression of the disease in patients with worsening RRMS, PRMS or SPMS; thus providing a new option for the management of these patients. The drug was generally well tolerated at the recommended dosage, although potential cardiotoxicity limits the total cumulative dose to 140 mg/m(2). Further studies are warranted to determine which patients with worsening RRMS, PRMS or SPMS are most likely to benefit from mitoxantrone treatment and to more fully define the long-term safety and tolerability of mitoxantrone, including the use of concomitant cardioprotectants to extend the therapeutic lifespan of the drug. Pharmacodynamic Profile. Mitoxantrone, a synthetic anthracenedione derivative, is an established cytotoxic, antineoplastic agent. Its presumed mechanism of action in multiple sclerosis (MS) is immunosuppression. In antineoplastic studies, the drug showed several immunomodulatory effects, inducing macrophage-mediated suppression of B-cell, T-helper and T-cytotoxic lymphocyte function. Currently, the pharmacodynamic properties of mitoxantrone have not been investigated to any extent in patients with MS. In one study, 6 months' treatment with intravenous mitoxantrone generally had no effect on the distribution of cytokine-positive peripheral blood monocyte cells in patients with MS. In an animal model of the disease, mitoxantrone suppressed the development and progression of both actively and passively induced acute experimental allergic encephalomyelitis (EAE). It appeared to be 10-20 times more effective than cyclophosphamide in the suppression of EAE. Moreover, mitoxantrone approximately doubled the mean time to onset of EAE versus control animals (279 vs 148 days after immunisation; p < 0.00005). In vitro, mitoxantrone 10 and 100 micro g/L inhibited myelin degradation by leucocytes and peritoneal macrophages derived from mice with acute EAE by approximately 60% and 100%. Pharmacokinetic Profile. Currently, there are no published pharmacokinetic data for intravenous mitoxantrone in pitoxantrone in patients with MS, paediatric patients or in those with renal impairment. All studies, to date, have been in patients with cancer receiving a single, approximately 30-minute intravenous infusion of mitoxantrone 5-14 mg/m(2). The drug exhibits triexponential pharmacokinetics, with a rapid initial distribution (alpha) phase, an intermediate distribution (beta) phase and a much slower elimination (gamma) phase. The mean half-life of the alpha phase appears to be 6-12 minutes and that of the beta phase 1.1-3.1 hours. Mitoxantrone has a high affinity for tissue, with a volume of distribution of up to 2248 L/m(2). Mitoxantrone persists for prolonged periods in tissues and was detectable in autopsy tissue from patients who last received the drug up to 272 days before death. At concentrations of 10-10000 ng/mL, the drug was 70-80 % bound to plasma proteins in dogs. Elimination of mitoxantrone occurs predominantly through biliary excretion and may be impaired in patients with hepatic dysfunction or third space abnormalities (e.g. ascites). The mean terminal elimination half-life of mitoxantrone ranged from 23 hours to 215 hours. Renal clearance accounts for 10 % of the total clearance of the drug. Total clearance of mitoxantrone ranged from 13 to 34.2 L/h/m(2) and renal clearance from 0.9 to 2.7 L/h/m(2). The drug appears to have a low potential for interaction with other concomitantly administered agents. Therapeutic Efficacy. Intravenous mitoxantrone (infusion of > or = 5 minutes), either as monotherapy or in combination with intravenous methylprednisolone, delayed the progression of the disease in patients with secondary-progressive (SP) or worsening relapsing-remitting (RR) MS (the latter is also termed progressive-relapsing MS) in comparative, randomised, multicentre trials. In a double-blind, monotherapy trial (Mitoxantrone In Multiple Sclerosis [MIMS] trial), mitoxantrone 12 mg/m(2) (n = 60) once every 3 months for 2 years significantly improved neurological disability relative to placebo (n = 64), as assessed by changes in mean Kurtzke Expanded Disability Status Scale (EDSS) score, mean Ambulatory Index (AI) score and mean Standardised Neurological Status (SNS) score. The drug also significantly reduced the mean number of corticosteroid-treated relapses per patient and prolonged the time to the first treated relapse. A Wei-Lachin multivariate analysis of these five efficacy variables indicated that the global difference between the two treatment groups was 0.30 (p < 0.0001). Mitroxantrone was also more effective than placebo according to secondary endpoints in this study, with fewer mitoxantrone recipients experiencing a relapse, a deterioration of > or =1 EDSS point or a confirmed deterioration in EDSS score over a 3-month period. Mitoxantrone recipients also showed less deterioration in quality-of-life ratings and had fewer hospital admissions, whereas more placebo recipients had new gadolinium-enhanced lesions at study end (the latter parameter was assessed using magnetic resonance imaging [MRI] in a subgroup of 110 patients, including 40 patients who received an exploratory 5 mg/m(2) dose). Furthermore, post hoc analyses indicated that the beneficial effects of mitoxantrone treatment on EDSS, SNS and AI scores were sustained for at least 12 months after cessation of treatment, with mean changes from baseline at 36 months in EDSS, AI and SNS scores of 0.10, 0.61 and 0.19, respectively, in the mitoxantrone group versus 0.46, 1.13 and 3.38 with placebo. Preliminary data from a cost-minimisation analysis based on results from the MIMS trial indicated that approximately half of the cost of mitoxantrone was offset by cost savings in other areas associated with the treatment of MS (direct and indirect major costs), with a total annual incremental cost for mitoxantrone of dollar 1661 per patient. Combination therapy once-monthly with intravenous mitoxantrone 20mg plus intravenous methylprednisolone 1g was more effective than intravenous methylprednisolone 1g once every month in preventing the development of gadolinium-enhanced lesions in patients with very active RRMS or SPMS (double-blind assessment using MRI scans). After 6 months, significantly more combination therapy recipients had no new gadolinium-enhanced lesions (90.5% vs 31.3% with monotherapy; p < 0.001) [primary endpoint]. There were also significant reductions in both the mean number of new enhancing lesions and the total number of gadolinium-enhanced lesions in patients receiving combination therapy versus methylprednisolone monotherapy.Tolerability. Mitoxantrone was generally well tolerated in patients with MS. Treatment-emergent adverse events occurring significantly more frequently with mitoxantrone (12 mg/m(2) once every 3 months for 2 years) than placebo were nausea, alopecia, menstrual disorders, urinary tract infection, amenorrhoea, leucopenia and elevated gamma-glutamyltranspeptidase levels. Adverse events were usually mild to moderate in severity and generally resolved with discontinuation of treatment or when treated with appropriate pharmacotherapy. Eight percent of patients discontinued treatment in the mitoxantrone 12 mg/m(2) group due to an adverse event versus 3% of placebo recipients. The incidence of drug-related acute myelogenous leukaemia was very low (0.12%) in a cohort of 802 patients with MS receiving mitoxantrone. Evidence suggests that the risk of cardiotoxicity is low in patients with MS. After 1 year of monotherapy, 3.4% of mitoxantrone recipients had a reduction in left ventricular ejection fraction (LVEF) to < or =50% compared with 0% of placebo recipients; at the end of the second year, respective incidences were 1.9% and 2.9% (total cumulative dose of mitoxantrone per patient was 96 mg/m(2) after 2 years' treatment). (ABSTRACT TRUNCATED)
Collapse
|
48
|
Wildiers H, Highley MS, de Bruijn EA, van Oosterom AT. Pharmacology of anticancer drugs in the elderly population. Clin Pharmacokinet 2004; 42:1213-42. [PMID: 14606930 DOI: 10.2165/00003088-200342140-00003] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Modifications to bodily functions and physiology are known to occur with age. These changes can have a considerable impact on the pharmacokinetic processes of absorption, distribution, metabolism and excretion and the pharmacodynamic properties of administered drugs. For many drugs with a high therapeutic index, this will be clinically unimportant, but for anticancer drugs, which usually have a low therapeutic index, these pharmacological changes can lead to dramatic consequences, such as excessive drug concentrations and unacceptable toxicity, or subtherapeutic drug concentrations and ineffective treatment. Despite the increased susceptibility of the elderly to these changes, doses are rarely adapted on the basis of pharmacokinetics and pharmacodynamics, with the exception of changes secondary to altered renal function. Until recently, only a few large prospective randomised trials have provided evidence-based data for dose adaptations in elderly patients. However, with increasing knowledge of the pharmacokinetics of anticancer drugs, advances in the knowledge of pharmacokinetic behaviour with aging, and documented efficacy and toxicity data in the elderly population, it is possible to highlight aspects of prescribing anticancer drugs in the elderly. In general, and for most drugs, age itself is not a contraindication to full-dose chemotherapy. The main limiting factors are comorbidity and poor functional status, which may be present in a significant number of the elderly population. Elderly patients with cancer are part of the daily practice of oncologists, but currently clinicians can often only estimate whether dose modification is advantageous for the elderly. This review attempts to elucidate the factors that can influence the pharmacokinetics of anticancer drugs frequently used in the elderly, and the clinical or biochemical parameters that form the basis for dose adjustments with age.
Collapse
Affiliation(s)
- Hans Wildiers
- Laboratory of Experimental Oncology, and Department of Medical Oncology, University Hospital Gasthuisberg, Leuven, Belgium.
| | | | | | | |
Collapse
|
49
|
Huang Y, Ghandehari H, Duan Y, Nan A, Zhang Z. HPMA copolymer-mitoxantrone conjugates for targeted cancer chemotherapy. J Drug Deliv Sci Technol 2004. [DOI: 10.1016/s1773-2247(04)50099-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Mross K, Scheulen ME, Licht T, Unger C, Richly H, Stern AC, Kutz K, Camboni MG, Barbieri P, Verdi E, Vincenzi B, Bernareggi A. Phase I clinical and pharmacokinetic study of BBR 3576, a novel aza-anthrapyrazole, administered i.v. every 4 weeks in patients with advanced solid tumors: a phase I study group trial of the Central European Society of Anticancer-Drug Research (CESAR). Anticancer Drugs 2004; 15:15-22. [PMID: 15090738 DOI: 10.1097/00001813-200401000-00003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BBR 3576 is a novel aza-anthrapyrazole with limited potential for cardiotoxicity in preclinical models. This phase I clinical and pharmacokinetic study was performed to determine the maximum tolerated dose, the dose-limiting toxicity (DLT) and the pharmacokinetic profile of BBR 3576 administered i.v. as a 1-h infusion repeated every 4 weeks. In total, 27 patients were treated at doses starting from 1 to 150 mg/m2. The dose levels 1, 2, 4, 8, 16, 32, 64, 90, 125 and 150 mg/m2 were investigated in one, three, one, three, two, one, three, four, three and six patients, respectively. The DLT was a grade 3 stomatitis at 150 mg/m2. At this dose level as well as at 125 mg/m2, neutropenia grade 3 and 4 were frequently seen, but not reaching the criteria for DLT. Time to neutrophil nadir was about 2 weeks and recovery took place within 1 week. Other bone marrow toxicities were mild; lymphopenia was also observed. No significant drug-induced cardiotoxicity was observed. The plasma concentration versus time curves of BBR 3576 showed a biexponential profile with a linear kinetic behavior. A very large volume of distribution, a high plasma clearance and long elimination half-lives were calculated. Renal unchanged drug excretion was less than 10% and therefore a minor excretion route. No objective antitumor responses were found. On the basis of this study, the recommended dose for phase II studies is 150 mg/m2, although the maximum tolerated dose as per protocol definition was not reached. This trial showed that BBR 3576 has a manageable toxicity profile on a 4-week schedule. Phase II studies have started in patients with solid tumors, as suggested by preclinical data in different in vivo model systems.
Collapse
Affiliation(s)
- Klaus Mross
- Tumor Biology Center Albert-Ludwigs University, Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|