1
|
Dmello C, Brenner A, Piccioni D, Wen PY, Drappatz J, Mrugala M, Lewis LD, Schiff D, Fadul CE, Chamberlain M, Kesari S, Ahluwalia M, Ghosh D, Sonabend AM, Kumthekar P. Phase II trial of blood-brain barrier permeable peptide-paclitaxel conjugate ANG1005 in patients with recurrent high-grade glioma. Neurooncol Adv 2024; 6:vdae186. [PMID: 39713041 PMCID: PMC11662161 DOI: 10.1093/noajnl/vdae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
Background This study is a phase II clinical trial to evaluate the efficacy, safety, and tolerability of the blood-brain barrier (BBB) permeable peptide-paclitaxel conjugate ANG1005 in patients with recurrent high-grade glioma (HGG) (NCT01967810). Methods Seventy-three patients were enrolled in 3 separate arms-recurrent glioblastoma (GBM) (Arm 1), bevacizumab refractory GBM (Arm 2), and grade 3 anaplastic gliomas (AGs) (Arm 3). The study was started in October 2013, and the data were locked on September 29, 2017. Safety was evaluated for all three arms (n = 73), and the primary endpoint for Arms 1 and 3 was objective response rate (ORR), and Arm 2 primary endpoint was progression-free survival rate at 3 months (PFS3). Results Overall, the safety of ANG1005 was found to be consistent with a taxane toxicity profile. Otherwise, the primary efficacy endpoints of ORR and PFS were not met. The most common adverse events (AEs) were hematologic (32.9%), alopecia (31.5%), and fatigue (30.1%). The median PFS was 1.4 months (95% CI: 1.4, 2.1) and similar across all the treatment arms. The median overall survival was 13.4 months (95% CI: 3.4, 14.6) in Arm 1, 5.8 months (95% CI: 1.9, 9.7) in Arm 2, and 18.2 months (95% CI: 10.7, 35.3) in Arm 3. Conclusion A dose of 600 mg/m2 was determined to be safe in this study. However, the primary efficacy endpoint was not met in the NCT01967810-ANG1005 trial, and no further studies are planned in the glioma setting with this compound.
Collapse
Affiliation(s)
- Crismita Dmello
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andrew Brenner
- Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas, USA
| | - David Piccioni
- Department of Neurosciences, UC San Diego Moores Cancer Center, La Jolla, California, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jan Drappatz
- Department of Neurology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Maciej Mrugala
- Fred Hutchinson Cancer Center, University of Washington Medical Center Seattle, Washington, USA
| | - Lionel D Lewis
- Department of Medicine, Dartmouth-Hitchcock Medical Center Lebanon, New Hampshire, USA
| | - David Schiff
- Emily Couric Clinical Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| | - Camilo E Fadul
- Division of Neuro-Oncology, Department of Neurology, University of Virginia, Charlottesville, Virginia, USA
| | | | - Santosh Kesari
- Department of Translational Neurosciences, Pacific Neuroscience Institute and Saint John’s Cancer Institute at Providence Saint John’s Health Center, Santa Monica, California, USA
| | - Manmeet Ahluwalia
- Department of Medical Oncology, Miami Cancer Institute, Miami, Florida, USA
| | - Debora Ghosh
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical Center, Boston, Massachusetts, USA
| | - Adam M Sonabend
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Priya Kumthekar
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Dong C, Yu X, Jin K, Qian J. Overcoming brain barriers through surface-functionalized liposomes for glioblastoma therapy; current status, challenges and future perspective. Nanomedicine (Lond) 2023; 18:2161-2184. [PMID: 38180008 DOI: 10.2217/nnm-2023-0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024] Open
Abstract
Glioblastoma (GB) originating from astrocytes is considered a grade IV astrocytoma tumor with severe consequences. The blood-brain barrier (BBB) offers a major obstacle in drug delivery to the brain to overcome GB. The current treatment options possess limited efficacy and maximal systemic toxic effects in GB therapy. Emerging techniques such as targeted drug delivery offer significant advantages, including enhanced drug delivery to the tumor site by overcoming the BBB. This review article focuses on the status of surface-modified lipid nanocarriers with functional ligands to efficiently traverse the BBB and improve brain targeting for successful GB treatment. The difficulties with surface-functionalized liposomes and potential future directions for opening up novel treatment options for GB are highlighted.
Collapse
Affiliation(s)
- Changming Dong
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Xuebin Yu
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, 312500, China
| |
Collapse
|
3
|
Straehla JP, Reardon DA, Wen PY, Agar NYR. The Blood-Brain Barrier: Implications for Experimental Cancer Therapeutics. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:265-289. [PMID: 38323268 PMCID: PMC10846865 DOI: 10.1146/annurev-cancerbio-061421-040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The blood-brain barrier is critically important for the treatment of both primary and metastatic cancers of the central nervous system (CNS). Clinical outcomes for patients with primary CNS tumors are poor and have not significantly improved in decades. As treatments for patients with extracranial solid tumors improve, the incidence of CNS metastases is on the rise due to suboptimal CNS exposure of otherwise systemically active agents. Despite state-of-the art surgical care and increasingly precise radiation therapy, clinical progress is limited by the ability to deliver an effective dose of a therapeutic agent to all cancerous cells. Given the tremendous heterogeneity of CNS cancers, both across cancer subtypes and within a single tumor, and the range of diverse therapies under investigation, a nuanced examination of CNS drug exposure is needed. With a shared goal, common vocabulary, and interdisciplinary collaboration, the field is poised for renewed progress in the treatment of CNS cancers.
Collapse
Affiliation(s)
- Joelle P Straehla
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Awuah WA, Kalmanovich J, Mehta A, Huang H, Abdul-Rahman T, Cheng Ng J, Yarlagadda R, Kamanousa K, Kundu M, Nansubuga EP, Hasan MM, Lyndin M, Isik A, Sikora V, Alexiou A. Multilevel Pharmacological Effects of Antipsychotics in Potential Glioblastoma Treatment. Curr Top Med Chem 2023; 23:389-402. [PMID: 36593538 DOI: 10.2174/1568026623666230102095836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 01/04/2023]
Abstract
Glioblastoma Multiforme (GBM) is a debilitating type of brain cancer with a high mortality rate. Despite current treatment options such as surgery, radiotherapy, and the use of temozolomide and bevacizumab, it is considered incurable. Various methods, such as drug repositioning, have been used to increase the number of available treatments. Drug repositioning is the use of FDA-approved drugs to treat other diseases. This is possible because the drugs used for this purpose have polypharmacological effects. This means that these medications can bind to multiple targets, resulting in multiple mechanisms of action. Antipsychotics are one type of drug used to treat GBM. Antipsychotics are a broad class of drugs that can be further subdivided into typical and atypical classes. Typical antipsychotics include chlorpromazine, trifluoperazine, and pimozide. This class of antipsychotics was developed early on and primarily works on dopamine D2 receptors, though it can also work on others. Olanzapine and Quetiapine are examples of atypical antipsychotics, a category that was created later. These medications have a high affinity for serotonin receptors such as 5- HT2, but they can also act on dopamine and H1 receptors. Antipsychotic medications, in the case of GBM, also have other effects that can affect multiple pathways due to their polypharmacological effects. These include NF-B suppression, cyclin deregulation, and -catenin phosphorylation, among others. This review will delve deeper into the polypharmacological, the multiple effects of antipsychotics in the treatment of GBM, and an outlook for the field's future progression.
Collapse
Affiliation(s)
| | | | - Aashna Mehta
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Helen Huang
- Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Jyi Cheng Ng
- Faculty of Medicine and Health Sciences, University of Putra Malaysia, Serdang, Malaysia
| | - Rohan Yarlagadda
- School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Karl Kamanousa
- College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Mrinmoy Kundu
- Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | | | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Mykola Lyndin
- Department of Medical Sciences, Sumy State University, Sumy, Ukraine
| | - Arda Isik
- Department of General Surgery, Istanbul Medeniyet University, Istanbul, Turkey
| | - Vladyslav Sikora
- Department of Medical Sciences, Sumy State University, Sumy, Ukraine
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
- AFNP Med, 1030 Wien, Austria
| |
Collapse
|
5
|
Zhang M, Yang C, Ruan X, Liu X, Wang D, Liu L, Shao L, Wang P, Dong W, Xue Y. CPEB2 m6A methylation regulates blood-tumor barrier permeability by regulating splicing factor SRSF5 stability. Commun Biol 2022; 5:908. [PMID: 36064747 PMCID: PMC9445078 DOI: 10.1038/s42003-022-03878-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
The blood–tumor barrier (BTB) contributes to poor therapeutic efficacy by limiting drug uptake; therefore, elevating BTB permeability is essential for glioma treatment. Here, we prepared astrocyte microvascular endothelial cells (ECs) and glioma microvascular ECs (GECs) as in vitro blood–brain barrier (BBB) and BTB models. Upregulation of METTL3 and IGF2BP3 in GECs increased the stability of CPEB2 mRNA through its m6A methylation. CPEB2 bound to and increased SRSF5 mRNA stability, which promoted the ETS1 exon inclusion. P51-ETS1 promoted the expression of ZO-1, occludin, and claudin-5 transcriptionally, thus regulating BTB permeability. Subsequent in vivo knockdown of these molecules in glioblastoma xenograft mice elevated BTB permeability, promoted doxorubicin penetration, and improved glioma-specific chemotherapeutic effects. These results provide a theoretical and experimental basis for epigenetic regulation of the BTB, as well as insight into comprehensive glioma treatment. The methyl transferase METTL3 is up-regulated in brain tumors leading to the methylation of CPEB2 mRNA, which in turn stabilizes the splicing factor SRSF5 mRNA, leading to the incorporation of exon 7 in ETS-1 in models of the Blood–Tumor Barrier.
Collapse
Affiliation(s)
- Mengyang Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, PR China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, PR China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, PR China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, PR China.,Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, PR China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, PR China
| | - Xuelei Ruan
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, PR China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, PR China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, PR China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, PR China.,Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, PR China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, PR China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, PR China.,Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, PR China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, PR China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, PR China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, PR China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, PR China
| | - Lianqi Shao
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, PR China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, PR China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, PR China
| | - Ping Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, PR China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, PR China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, PR China
| | - Weiwei Dong
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, PR China.,Liaoning Research Center for Translational Medicine in Nervous System Disease, Shenyang, PR China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, PR China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, PR China. .,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, PR China. .,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, PR China.
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Autoimmune encephalitis (AE) refers to immune-mediated neurological syndromes often characterised by the detection of pathogenic autoantibodies in serum and/or cerebrospinal fluid which target extracellular epitopes of neuroglial antigens. There is increasing evidence these autoantibodies directly modulate function of their antigens in vivo. Early treatment with immunotherapy improves outcomes. Yet, these patients commonly exhibit chronic disability. Importantly, optimal therapeutic strategies at onset and during escalation remain poorly understood. In this review of a rapidly emerging field, we evaluate recent studies on larger cohorts, registries, and meta-analyses to highlight existing evidence for contemporary therapeutic approaches in AE. RECENT FINDINGS We highlight acute and long-term treatments used in specific AE syndromes, exemplify how understanding disease pathogenesis can inform precision therapy and outline challenges of defining disability outcomes in AE. SUMMARY Early first-line immunotherapies, including corticosteroids and plasma exchange, improve outcomes, with emerging evidence showing second-line immunotherapies (especially rituximab) reduce relapse rates. Optimal timing of immunotherapy escalation remains unclear. Routine reporting of outcome measures which incorporate cognitive impairment, fatigue, pain, and mental health will permit more accurate quantification of residual disability and comprehensive comparisons between international multicentre cohorts, and enable future meta-analyses with the aim of developing evidence-based therapeutic guidelines.
Collapse
Affiliation(s)
- Benjamin P Trewin
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead; Sydney Medical School and Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Isaak Freeman
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sudarshini Ramanathan
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead; Sydney Medical School and Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Department of Neurology, Concord Hospital, Sydney, Australia
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK
| |
Collapse
|
7
|
Development and Validation of an HPLC Method for Analysis of Topotecan in Human Cerebrospinal Fluid and Its Application in Elimination Evaluation of Topotecan after Intraventricular Injection. Cancers (Basel) 2021; 13:cancers13184643. [PMID: 34572867 PMCID: PMC8471976 DOI: 10.3390/cancers13184643] [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: 08/06/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
Intrathecal administration of anticancer drugs is an effective dosage strategy, but the elimination of intraventricular drugs is not uniform in all patients. For safety, a system to evaluate local pharmacokinetics in the ventricles after administration is desired. In this study, we developed a simple and reproducible method to measure topotecan concentration in the cerebrospinal fluid (CSF) and confirmed its clinical applicability. High-performance liquid chromatography (HPLC) analysis was performed using a C18 column to measure the total topotecan concentration in the CSF. Clinical CSF samples were obtained from a 1-year old child with poor CSF absorption and stagnation. The patient received topotecan via an intraventricular subcutaneous reservoir. The HPLC method complied with the validation criteria. The lower limit of quantitation of this method was 0.04 µM. Using the developed method, we could determine the difference in topotecan CSF concentrations at 24 and 48 h after administration. The patient's topotecan elimination rate was extremely low, and signs of adverse effects were observed at high CSF concentration of topotecan. The developed method could detect the delay in topotecan elimination after intrathecal injection. The findings of this study are valuable for the development of personalized treatments for the intrathecal administration of anticancer drugs.
Collapse
|
8
|
Augustine R, Aqel AH, Kalva SN, Joshy KS, Nayeem A, Hasan A. Bioengineered microfluidic blood-brain barrier models in oncology research. Transl Oncol 2021; 14:101087. [PMID: 33865030 PMCID: PMC8066424 DOI: 10.1016/j.tranon.2021.101087] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/25/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Metastasis is the major reason for most brain tumors with up to a 50% chance of occurrence in patients with other types of malignancies. Brain metastasis occurs if cancer cells succeed to cross the 'blood-brain barrier' (BBB). Moreover, changes in the structure and function of BBB can lead to the onset and progression of diseases including neurological disorders and brain-metastases. Generating BBB models with structural and functional features of intact BBB is highly important to better understand the molecular mechanism of such ailments and finding novel therapeutic agents targeting them. Hence, researchers are developing novel in vitro BBB platforms that can recapitulate the structural and functional characteristics of BBB. Brain endothelial cells-based in vitro BBB models have thus been developed to investigate the mechanism of brain metastasis through BBB and facilitate the testing of brain targeted anticancer drugs. Bioengineered constructs integrated with microfluidic platforms are vital tools for recapitulating the features of BBB in vitro closely as possible. In this review, we outline the fundamentals of BBB biology, recent developments in the microfluidic BBB platforms, and provide a concise discussion of diverse types of bioengineered BBB models with an emphasis on the application of them in brain metastasis and cancer research in general. We also provide insights into the challenges and prospects of the current bioengineered microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| | - Ahmad H Aqel
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Sumama Nuthana Kalva
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - K S Joshy
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur 680020, Kerala, India
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| |
Collapse
|
9
|
Cancela MB, Zugbi S, Winter U, Martinez AL, Sampor C, Sgroi M, Francis JH, Garippa R, Abramson DH, Chantada G, Schaiquevich P. A decision process for drug discovery in retinoblastoma. Invest New Drugs 2020; 39:426-441. [PMID: 33200242 DOI: 10.1007/s10637-020-01030-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/28/2020] [Indexed: 11/28/2022]
Abstract
Intraocular retinoblastoma treatment has changed radically over the last decade, leading to a notable improvement in ocular survival. However, eyes that relapse remain difficult to treat, as few alternative active drugs are available. More challenging is the scenario of central nervous system (CNS) metastasis, in which almost no advancements have been made. Both clinical scenarios represent an urgent need for new drugs. Using an integrated multidisciplinary approach, we developed a decision process for prioritizing drug selection for local (intravitreal [IVi], intrathecal/intraventricular [IT/IVt]), systemic, or intra-arterial chemotherapy (IAC) treatment by means of high-throughput pharmacological screening of primary cells from two patients with intraocular tumor and CNS metastasis and a thorough database search to identify clinical and biopharmaceutical data. This process identified 169 compounds to be cytotoxic; only 8 are FDA-approved, lack serious toxicities and available for IVi administration. Four of these agents could also be delivered by IT/IVt. Twelve FDA-approved drugs were identified for systemic delivery as they are able to cross the blood-brain barrier and lack serious adverse events; four drugs are of oral usage and six compounds that lack vesicant or neurotoxicity could be delivered by IAC. We also identified promising compounds in preliminary phases of drug development including inhibitors of survivin, antiapoptotic Bcl-2 family proteins, methyltransferase, and kinesin proteins. This systematic approach may be applied more broadly to prioritize drugs to be repurposed or to identify novel hits for use in retinoblastoma treatment.
Collapse
Affiliation(s)
- María Belen Cancela
- Precision Medicine, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina.,National Scientific and Technical Research Council, CONICET, 1425, Buenos Aires, Argentina
| | - Santiago Zugbi
- Precision Medicine, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina.,National Scientific and Technical Research Council, CONICET, 1425, Buenos Aires, Argentina
| | - Ursula Winter
- Pathology Service, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina
| | - Ana Laura Martinez
- Precision Medicine, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina
| | - Claudia Sampor
- Hematology-Oncology Service, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina
| | - Mariana Sgroi
- Ophthalmology Service, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina
| | - Jasmine H Francis
- Ophthalmic Oncology Service, Memorial Sloan-Kettering Institute and Cancer Center, New York, NY, 10065, USA
| | - Ralph Garippa
- Gene Editing And Screening Core facility, Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Institute and Cancer Center, New York, NY, 10065, USA
| | - David H Abramson
- Ophthalmic Oncology Service, Memorial Sloan-Kettering Institute and Cancer Center, New York, NY, 10065, USA
| | - Guillermo Chantada
- Precision Medicine, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina.,National Scientific and Technical Research Council, CONICET, 1425, Buenos Aires, Argentina
| | - Paula Schaiquevich
- Precision Medicine, Hospital de Pediatría JP Garrahan, 1245, Buenos Aires, Argentina. .,National Scientific and Technical Research Council, CONICET, 1425, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Yoon WS, Park JS, Kim YI, Chung DS, Jeun SS, Hong YK, Yang SH. High-dose methotrexate monotherapy for newly diagnosed primary central nervous system lymphoma: 15-year multicenter experience. Asia Pac J Clin Oncol 2020; 17:123-130. [PMID: 32978898 DOI: 10.1111/ajco.13427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 06/20/2020] [Indexed: 12/23/2022]
Abstract
AIM Primary central nervous system lymphoma (PCNSL) is rare disease and shows poor prognosis although methotrexate-based chemotherapy is used. Here, we present our experiences with high-dose methotrexate (HD-MTX) monotherapy for immunocompetent patients with PCNSL at three institutions and investigate factors related to survival. METHODS PCNSL patients, who were histologically confirmed with diffuse large B cells and treated with HD-MTX monotherapy from 2001 to 2016, were retrospectively reviewed. Patients underwent induction chemotherapy with 8 g/m2 of MTX every 10 days (maximum three cycles). Maintenance chemotherapy of 3.5 g/m2 of MTX (maximum six cycles) was selectively performed depending on the response to induction chemotherapy. RESULTS A total of 67 patients were included. Although seven patients discontinued induction chemotherapy because of MTX toxicity, 40 (59.7%) patients showed a complete response (CR) to induction chemotherapy. Twenty-six (38.8%) and three (4.5%) patients showed a CR and partial response, respectively, after maintenance chemotherapy. Forty-one patients with recurrence or progression following HD-MTX underwent second-line treatment. Progression-free survival rates were 43% and 24% at 1 and 2 years, respectively. The median overall survival was 40.3 months. In a multivariate analysis, a radiological CR to induction chemotherapy was a significant factor related to prolonged progression-free survival and overall survival (P < 0.05). CONCLUSION MTX-monotherapy is tolerable in terms of adverse effects and still considered as a treatment option in patients with PCNSL. However, an additional therapeutic option should be prepared for non-CR responders to induction chemotherapy.
Collapse
Affiliation(s)
- Wan-Soo Yoon
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Jae-Sung Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young-Il Kim
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Dong-Sup Chung
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Kil Hong
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent's Hospital, Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Suwon, Korea
| |
Collapse
|
11
|
Longuespée R, Theile D, Fresnais M, Burhenne J, Weiss J, Haefeli WE. Approaching sites of action of drugs in clinical pharmacology: New analytical options and their challenges. Br J Clin Pharmacol 2020; 87:858-874. [PMID: 32881012 DOI: 10.1111/bcp.14543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Clinical pharmacology is an important discipline for drug development aiming to define pharmacokinetics (PK), pharmacodynamics (PD) and optimum exposure to drugs, i.e. the concentration-response relationship and its modulators. For this purpose, information on drug concentrations at the anatomical, cellular and molecular sites of action is particularly valuable. In pharmacological assays, the limited accessibility of target cells in readily available samples (i.e. blood) often hampers mass spectrometry-based monitoring of the absolute quantity of a compound and the determination of its molecular action at the cellular level. Recently, new sample collection methods have been developed for the specific capture of rare circulating cells, especially for the diagnosis of circulating tumour cells. In parallel, new advances and developments in mass spectrometric instrumentation now allow analyses to be scaled down to the cellular level. Together, these developments may permit the monitoring of minute drug quantities and show their effect at the cellular level. In turn, such PK/PD associations on a cellular level would not only enrich our pharmacological knowledge of a given compound but also expand the basis for PK/PD simulations. In this review, we describe novel concepts supporting clinical pharmacology at the anatomical, cellular and molecular sites of action, and highlight the new challenges in mass spectrometry-based monitoring. Moreover, we present methods to tackle these challenges and define future needs.
Collapse
Affiliation(s)
- Rémi Longuespée
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Dirk Theile
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Margaux Fresnais
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany.,German Cancer Consortium (DKTK)-German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
12
|
Preventive hypothermia as a neuroprotective strategy for paclitaxel-induced peripheral neuropathy. Pain 2020; 160:1505-1521. [PMID: 30839425 DOI: 10.1097/j.pain.0000000000001547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a severe adverse effect that occurs secondary to anticancer treatments and has no known preventive or therapeutic strategy. Therapeutic hypothermia has been shown to be effective in protecting against central and peripheral nervous system injuries. However, the effects of therapeutic hypothermia on CIPN have rarely been explored. We induced lower back hypothermia (LBH) in an established paclitaxel-induced CIPN rat model and found that the paclitaxel-induced impairments observed in behavioral, electrophysiological, and histological impairments were inhibited by LBH when applied at an optimal setting of 24°C to the sciatic nerve and initiated 90 minutes before paclitaxel infusion. Lower back hypothermia also inhibited the paclitaxel-induced activation of astroglia and microglia in the spinal cord and macrophage infiltration into and neuronal injury in the dorsal root ganglia and sciatic nerves. Furthermore, LBH decreased the local blood flow and local tissue concentrations of paclitaxel. Finally, in NOD/SCID mice inoculated with cancer cells, the antiproliferative effect of paclitaxel was not affected by the distal application of LBH. In conclusion, our findings indicate that early exposure to regional hypothermia alleviates paclitaxel-induced peripheral neuropathy. Therapeutic hypothermia may therefore represent an economical and nonpharmaceutical preventive strategy for CIPN in patients with localized solid tumors.
Collapse
|
13
|
Hyung J, Hong JY, Yoon DH, Kim S, Park JS, Park CS, Lee SW, Kim JH, Ryu JS, Huh J, Suh C. Thiotepa, busulfan, and cyclophosphamide or busulfan, cyclophosphamide, and etoposide high-dose chemotherapy followed by autologous stem cell transplantation for consolidation of primary central nervous system lymphoma. Ann Hematol 2019; 98:1657-1664. [PMID: 30989325 DOI: 10.1007/s00277-019-03667-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/07/2019] [Indexed: 10/27/2022]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare extranodal non-Hodgkin lymphoma for which standard treatment has yet to be established. High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) is a suitable consolidation strategy for patients who respond to induction chemotherapy. The purpose of this study was to compare the outcome and toxicity profile of the combination of busulfan, cyclophosphamide, and etoposide (BuCyE) with that of the combination of thiotepa, busulfan, and cyclophosphamide (TBC) as conditioning regimens of upfront ASCT for consolidation therapy in PCNSL. The PCNSL registry data set, prospectively collected from March 1993 to May 2017 at Asan Medical Center, was reviewed retrospectively. Patients with objective response to induction chemotherapy who received BuCyE or TBC as conditioning regimen for ASCT were included in the analysis. Primary endpoints were overall survival (OS) and progression-free survival (PFS). Among 241 patients with a diagnosis of PCNSL, 53 received ASCT as upfront consolidation therapy with TBC (28 patients) or BuCyE (25 patients) as conditioning regimen. No median OS or PFS was reached in the TBC group, while the BuCyE group reached a median OS of 4.9 years (p = 0.02) and median PFS of 1.1 years (p = 0.007). The incidence of oral mucositis, nausea, and vomiting was higher with TBC than BuCyE. The median admission duration and days to engraftment were similar between the two groups. Despite the greater incidence of adverse events, TBC showed better outcomes than BuCyE in terms of survival.
Collapse
Affiliation(s)
- Jaewon Hyung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jung Yong Hong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Dok Hyun Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Shin Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jung Sun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Chan-Sik Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jeong Hoon Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jin Sook Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jooryung Huh
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Cheolwon Suh
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
14
|
Gaut D, Schiller GJ. Hematopoietic stem cell transplantation in primary central nervous system lymphoma: a review of the literature. Int J Hematol 2019; 109:260-277. [PMID: 30671909 DOI: 10.1007/s12185-019-02594-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/14/2018] [Accepted: 01/08/2019] [Indexed: 12/26/2022]
Abstract
Primary central nervous system lymphoma (PCNSL) is an uncommon variant of extranodal non-Hodgkin lymphoma (NHL) with an aggressive course and worse outcomes compared with other lymphomas of similar tumor burden and histologic subtype. High-dose chemotherapy supported by autologous stem cell transplantation (HDC/ASCT) is an option for therapy for this disease in both the relapse setting and as post-remission consolidation. Data are currently limited to only several single-arm phase II trials with small sample sizes, but randomized trials are now ongoing. In this review, we discuss the efficacy, feasibility, and toxicity of HDC/ASCT for PCNSL and its role in the treatment of this aggressive malignancy, both in the first-line and relapse settings. We also bring to attention the current data on allogeneic stem cell transplantation (allo-SCT) in PCNSL.
Collapse
Affiliation(s)
- Daria Gaut
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gary J Schiller
- Hematological Malignancies/Stem Cell Transplantation Unit, Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Room 42-121 CHS, Los Angeles, CA, 90095, USA.
| |
Collapse
|
15
|
Lee HM, Clark EP, Kuijer MB, Cushman M, Pommier Y, Philpot BD. Characterization and structure-activity relationships of indenoisoquinoline-derived topoisomerase I inhibitors in unsilencing the dormant Ube3a gene associated with Angelman syndrome. Mol Autism 2018; 9:45. [PMID: 30140420 PMCID: PMC6098585 DOI: 10.1186/s13229-018-0228-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/06/2018] [Indexed: 01/26/2023] Open
Abstract
Background Angelman syndrome (AS) is a severe neurodevelopmental disorder lacking effective therapies. AS is caused by mutations in ubiquitin protein ligase E3A (UBE3A), which is genomically imprinted such that only the maternally inherited copy is expressed in neurons. We previously demonstrated that topoisomerase I (Top1) inhibitors could successfully reactivate the dormant paternal allele of Ube3a in neurons of a mouse model of AS. We also previously showed that one such Top1 inhibitor, topotecan, could unsilence paternal UBE3A in induced pluripotent stem cell-derived neurons from individuals with AS. Although topotecan has been well-studied and is FDA-approved for cancer therapy, its limited CNS bioavailability will likely restrict the therapeutic use of topotecan in AS. The goal of this study was to identify additional Top1 inhibitors with similar efficacy as topotecan, with the expectation that these could be tested in the future for safety and CNS bioavailability to assess their potential as AS therapeutics. Methods We tested 13 indenoisoquinoline-derived Top1 inhibitors to identify compounds that unsilence the paternal allele of Ube3a in mouse neurons. Primary cortical neurons were isolated from embryonic day 14.5 (E14.5) mice with a Ube3a-YFP fluorescent tag on the paternal allele (Ube3am+/pYFP mice) or mice that lack the maternal Ube3a allele and hence model AS (Ube3am-/p+ mice). Neurons were cultured for 7 days, treated with drug for 72 h, and examined for paternal UBE3A protein expression by Western blot or fluorescence immunostaining. Dose responses of the compounds were determined across a log range of drug treatments, and cytotoxicity was tested using a luciferase-based assay. Results All 13 indenoisoquinoline-derived Top1 inhibitors unsilenced paternal Ube3a. Several compounds exhibited favorable paternal Ube3a unsilencing properties, similar to topotecan, and of these, indotecan (LMP400) was the most effective based on estimated Emax (maximum response of unsilencing paternal Ube3a) and EC50 (half maximal effective concentration). Conclusions We provide pharmacological profiles of indenoisoquinoline-derived Top1 inhibitors as paternal Ube3a unsilencers. All 13 tested compounds were effective at unsilencing paternal Ube3a, although with variable efficacy and potency. Indotecan (LMP400) demonstrated a better pharmacological profile of Ube3a unsilencing compared to our previous lead compound, topotecan. Taken together, indotecan and its structural analogues are potential AS therapeutics whose translational potential in AS treatment should be further assessed.
Collapse
Affiliation(s)
- Hyeong-Min Lee
- 1Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Neuroscience Research Building, Room 5119 115 Mason Farm Rd., Campus Box 7545, Chapel Hill, NC 27599-7545 USA
| | - Ellen P Clark
- 1Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Neuroscience Research Building, Room 5119 115 Mason Farm Rd., Campus Box 7545, Chapel Hill, NC 27599-7545 USA
| | - M Bram Kuijer
- 1Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Neuroscience Research Building, Room 5119 115 Mason Farm Rd., Campus Box 7545, Chapel Hill, NC 27599-7545 USA
| | - Mark Cushman
- 2Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University School of Pharmacy and the Purdue Center for Cancer Research, West Lafayette, IN USA
| | - Yves Pommier
- 3Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD USA
| | - Benjamin D Philpot
- 1Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Neuroscience Research Building, Room 5119 115 Mason Farm Rd., Campus Box 7545, Chapel Hill, NC 27599-7545 USA.,4UNC Neuroscience Center, Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC USA
| |
Collapse
|
16
|
Clinical benefit of continuing crizotinib therapy after initial disease progression in Chinese patients with advanced ALK-rearranged non-small-cell lung cancer. Oncotarget 2018; 8:41631-41640. [PMID: 28427213 PMCID: PMC5522304 DOI: 10.18632/oncotarget.15892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/22/2017] [Indexed: 11/25/2022] Open
Abstract
Purpose Although most patients with ALK-positive non?small-cell lung cancer (NSCLC) who benefit from treatment with crizotinib ultimately develop progressive disease (PD), continuing crizotinb beyond the initial PD (CBPD) in these patients may be beneficial. In this study, we investigated whether Chinese patients with advanced ALK-positive NSCLC benefit from CBPD, and whether any factors are predictive of a longer post-initial progression-free survival time (PFS2). Materials and Methods Data on 33 patients with ALK-positive NSCLC who achieved disease control with crizotinib were analyzed retrospectively. The impact of continued crizotinib therapy on the patients’ PFS2 time was assessed after adjusting for potential confounding factors. Results With initial crizotinib therapy, the objective response rate (ORR) and median PFS time (PFS1) in the 33 patients were 63.6% and 8.6 months, respectively. With continued crizotinib therapy after documentation of PD, the median PFS2 for all 33 patients was 16 weeks, and in those with CNS progression but systemic disease control it was 30 weeks. Patients who received local therapy after disease progression had a significantly longer PFS2 compared with those who did not (P = 0.039). Multivariable Cox regression analysis showed that the PFS1 with initial crizotinib treatment and local therapy were independent predictors of PFS2. Discussion This study provides further evidence of the benefit of continuing crizotinib therapy in Chinese patients with progressive ALK-positive NSCLC. Patients with a longer PFS1 and those who received local brain therapy would have a longer period of continuing crizotinib.
Collapse
|
17
|
Figueira TN, Oliveira FD, Almeida I, Mello ÉO, Gomes VM, Castanho MARB, Gaspar D. Challenging metastatic breast cancer with the natural defensin PvD 1. NANOSCALE 2017; 9:16887-16899. [PMID: 29076508 DOI: 10.1039/c7nr05872a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Metastatic breast cancer is a very serious life threatening condition that poses many challenges for the pharmaceutical development of effective chemotherapeutics. As the therapeutics targeted to the localized masses in breast improve, metastatic lesions in the brain slowly increase in their incidence compromising successful treatment outcomes overall. The blood-brain-barrier (BBB) is one important obstacle for the management of breast cancer brain metastases. New therapeutic approaches are in demand for overcoming the BBB's breaching by breast tumor cells. In this work we demonstrate the potential dual role of a natural antimicrobial plant defensin, PvD1: it interferes with the formation of solid tumors in the breast and concomitantly controls adhesion of breast cancer cells to human brain endothelial cells. We have used a combination of techniques that probe PvD1's effect at the single cell level and reveal that this peptide can effectively damage breast tumor cells, leaving healthy breast and brain cells unaffected. Results suggest that PvD1 quickly internalizes in cancer cells but remains located in the membrane of normal cells with no significant damage to its structure and biomechanical properties. These interactions in turn modulate cell adhesiveness between tumor and BBB cells. PvD1 is a potential template for the design of innovative pharmacological approaches for metastatic breast cancer treatment: the manipulation of the biomechanical properties of tumor cells that ultimately prevent their attachment to the BBB.
Collapse
Affiliation(s)
- Tiago N Figueira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon 1649-028, Portugal.
| | - Filipa D Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon 1649-028, Portugal.
| | - Inês Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon 1649-028, Portugal.
| | - Érica O Mello
- Laboratório de Fisiologia e Bioquímica de Microrganismos do Centro de Biociências e Biotecnologia da Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego 2000, Campos dos Goytacazes, Rio de Janeiro, 28013-602, Brazil
| | - Valdirene M Gomes
- Laboratório de Fisiologia e Bioquímica de Microrganismos do Centro de Biociências e Biotecnologia da Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego 2000, Campos dos Goytacazes, Rio de Janeiro, 28013-602, Brazil
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon 1649-028, Portugal.
| | - Diana Gaspar
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon 1649-028, Portugal.
| |
Collapse
|
18
|
Sun L, Joh DY, Al-Zaki A, Stangl M, Murty S, Davis JJ, Baumann BC, Alonso-Basanta M, Kaol GD, Tsourkas A, Dorsey JF. Theranostic Application of Mixed Gold and Superparamagnetic Iron Oxide Nanoparticle Micelles in Glioblastoma Multiforme. J Biomed Nanotechnol 2016; 12:347-56. [PMID: 27305768 DOI: 10.1166/jbn.2016.2173] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The treatment of glioblastoma multiforme, the most prevalent and lethal form of brain cancer in humans, has been limited in part by poor delivery of drugs through the blood-brain barrier and by unclear delineation of the extent of infiltrating tumor margins. Nanoparticles, which selectively accumulate in tumor tissue due to their leaky vasculature and the enhanced permeability and retention effect, have shown promise as both therapeutic and diagnostic agents for brain tumors. In particular, superparamagnetic iron oxide nanoparticles (SPIONs) have been leveraged as T2-weighted MRI contrast agents for tumor detection and imaging; and gold nanoparticles (AuNP) have been demonstrated as radiosensitizers capable of propagating electron and free radical-induced radiation damage to tumor cells. In this study, we investigated the potential applications of novel gold and SPION-loaded micelles (GSMs) coated by polyethylene glycol-polycaprolactone (PEG-PCL) polymer. By quantifying gh2ax DNA damage foci in glioblastoma cell lines, we tested the radiosensitizing efficacy of these GSMs, and found that GSM administration in conjunction with radiation therapy (RT) led to ~2-fold increase in density of double-stranded DNA breaks. For imaging, we used GSMs as a contrast agent for both computed tomography (CT) and magnetic resonance imaging (MRI) studies of stereotactically implanted GBM tumors in a mouse model, and found that MRI but not CT was sufficiently sensitive to detect and delineate tumor borders after administration and accumulation of GSMs. These results suggest that with further development and testing, GSMs may potentially be integrated into both imaging and treatment of brain tumors, serving a theranostic purpose as both an MRI-based contrast agent and a radiosensitizer.
Collapse
|
19
|
Fröscher W, Kirschstein T, Rösche J. Anticonvulsant therapy in brain-tumor related epilepsy. JOURNAL OF EPILEPTOLOGY 2016. [DOI: 10.1515/joepi-2016-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
SummaryBackground. The lifetime risk of patients with brain tumors to have focal epileptic seizures is 10-100%; the risk depends on different histology. Specific guidelines for drug treatment of brain tumor-related seizures have not yet been established.Aim. This review addresses the special aspects of antiepileptic drug (AED) therapy in brain tumor-related epilepsy.Methods. We analyzed the literature up to December 2015.Results. Based on current evidence the management of tumor-related seizures does not differ substantially from that applied to epilepsies from other etiologies. Therefore, the choice of an AED is based, above all, on tolerability and pharmacokinetic interactions with chemotherapeutic drugs. Levetiracetam is recommended by many authors as first-line therapy in brain tumor-related epilepsy. Due to the possibility of interactions, the combination of enzyme-inducing AEDs and chemotherapeutic drugs, is usually not recommended as a first choice. Currently there is no evidence that prophylactic prescription of long-term AEDs in brain tumor-patients who did not present with seizures is justified. Because of the high risk of recurrence, however, AED treatment should be strongly considered after a single brain tumor-related seizure. The decision to withdraw AEDs must carefully consider the risk of seizure recurrence.Conclusion. At present levetiracetam is the preferred drug in brain tumor-related epilepsy, especially when drug interactions need to be avoided. In the future we hope to acquire more targeted drugs against this disorder by uncovering its pathogenesis.
Collapse
|
20
|
da Ros M, Iorio AL, Lucchesi M, Stival A, de Martino M, Sardi I. The Use of Anthracyclines for Therapy of CNS Tumors. Anticancer Agents Med Chem 2016; 15:721-7. [PMID: 25846760 PMCID: PMC4997942 DOI: 10.2174/1871520615666150407155319] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 03/26/2015] [Accepted: 04/05/2015] [Indexed: 02/07/2023]
Abstract
Despite being long lived, anthracyclines remain the “evergreen” drugs in clinical practice of oncology, showing a potent effect in inhibiting cell growth in many types of tumors, including brain neoplasms. Unfortunately, they suffer from a poor penetration into the brain when intravenously administered due to multidrug resistance mechanism, which hampers their delivery across the blood brain barrier. In this paper, we summarize the current literature on the role of anthracyclines in cancer therapy and highlight recent efforts on 1) development of tumor cell resistance to anthracyclines and 2) the new approaches to brain drug delivery across the blood brain barrier.
Collapse
Affiliation(s)
| | | | | | | | | | - Iacopo Sardi
- Neuro-Oncology Unit, Department of Paediatric Medicine, Meyer Children's Hospital. Viale G. Pieraccini 24, 50139 Florence, Italy.
| |
Collapse
|
21
|
Jacus MO, Daryani VM, Harstead KE, Patel YT, Throm SL, Stewart CF. Pharmacokinetic Properties of Anticancer Agents for the Treatment of Central Nervous System Tumors: Update of the Literature. Clin Pharmacokinet 2016; 55:297-311. [PMID: 26293618 PMCID: PMC4761278 DOI: 10.1007/s40262-015-0319-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite significant improvement in outcomes for patients with hematologic malignancies and solid tumors over the past 10 years, patients with primary or metastatic brain tumors continue to have a poor prognosis. A primary reason for this is the inability of many chemotherapeutic drugs to penetrate into the brain and brain tumors at concentrations high enough to exert an antitumor effect because of unique barriers and efflux transporters. Several studies have been published recently examining the central nervous system pharmacokinetics of various anticancer drugs in patients with primary and metastatic brain tumors. To summarize recent advances in the field, this review critically presents studies published within the last 9 years examining brain and cerebrospinal fluid penetration of clinically available anticancer agents for patients with central nervous system tumors.
Collapse
Affiliation(s)
- Megan O Jacus
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Vinay M Daryani
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - K Elaine Harstead
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yogesh T Patel
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Stacy L Throm
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
22
|
Sullivan I, Planchard D. ALK inhibitors in non-small cell lung cancer: the latest evidence and developments. Ther Adv Med Oncol 2016; 8:32-47. [PMID: 26753004 PMCID: PMC4699265 DOI: 10.1177/1758834015617355] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The treatment of patients with advanced non-small cell lung cancer (NSCLC) harbouring chromosomal rearrangements of ALK (anaplastic lymphoma kinase) was revolutionized by crizotinib, a small molecule inhibitor of ALK, ROS1 and MET. Unfortunately, the disease progressed within the first 12 months in most of the patients because of the development of crizotinib resistance in the majority of patients and the emergence of acquired resistance mutations in most of them. Many of them had been reported even before its approval leading to the rapid development of second-generation ALK inhibitors for crizotinib-resistant NSCLC. In the last few years, novel potent ALK inhibitors with promising results and a good toxicity profile have become available: ceritinib (LDK378), alectinib (RG7853/AF-802/RO5424802/CH5424802), brigatinib (AP26113), entrectinib (RXDX-101, NMS-E628), PF-06463922, ASP3026, TSR-011, X-376/X-396 and CEP-28122/CEP-37440. Moreover, HSP90 (90 kDa heat shock protein) inhibitors have demonstrated clinical activity in patients with ALK+ NSCLC. This review focuses on the molecular and clinical properties of this new generation of ALK inhibitors under development in the clinic.
Collapse
Affiliation(s)
| | - David Planchard
- Gustave Roussy – Medical Oncology, 114 rue Édouard Vaillant, Villejuif 94805, France
| |
Collapse
|
23
|
Ferreri AJ, Donadoni G, Cabras MG, Patti C, Mian M, Zambello R, Tarella C, Di Nicola M, D'Arco AM, Doa G, Bruno-Ventre M, Assanelli A, Foppoli M, Citterio G, Fanni A, Mulè A, Caligaris-Cappio F, Ciceri F. High Doses of Antimetabolites Followed by High-Dose Sequential Chemoimmunotherapy and Autologous Stem-Cell Transplantation in Patients With Systemic B-Cell Lymphoma and Secondary CNS Involvement: Final Results of a Multicenter Phase II Trial. J Clin Oncol 2015; 33:3903-10. [DOI: 10.1200/jco.2015.61.1236] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Treatment of secondary CNS dissemination in patients with aggressive lymphomas remains an important, unmet clinical need. Herein, we report the final results of a multicenter phase II trial addressing a new treatment for secondary CNS lymphoma based on encouraging experiences with high doses of antimetabolites in primary CNS lymphoma and with rituximab plus high-dose sequential chemoimmunotherapy (R-HDS) in relapsed aggressive lymphoma. Patients and Methods HIV-negative patients with aggressive B-cell lymphoma and secondary CNS involvement at diagnosis or relapse, age 18 to 70 years, and Eastern Cooperative Oncology Group performance status ≤ 3 were enrolled and treated with high-doses of methotrexate and cytarabine, followed by R-HDS (cyclophosphamide, cytarabine, and etoposide) supported by autologous stem-cell transplantation (ASCT). Treatment included eight doses of rituximab and four doses of intrathecal liposomal cytarabine. The primary end point was 2-year event-free survival; the planned accrual was 38 patients. Results Thirty-eight patients were enrolled; CNS disease was detected at presentation in 16 patients. Toxicity was usually hematologic and manageable, with grade 4 febrile neutropenia in 3% of delivered courses and grade 4 nonhematologic toxicity in 2% of delivered courses. Four patients died because of toxicity. Autologous stem cells were successfully collected in 24 (89%) of 27 patients (median, 10 × 106/kg); 20 patients underwent ASCT. Complete response was achieved in 24 patients (complete response rate, 63%; 95% CI, 48% to 78%). At a median follow-up of 48 months, 17 patients remained relapse free, with a 2-year event-free survival rate of 50% ± 8%. At 5 years, 16 patients were alive, with a 5-year overall survival rate of 41% ± 8% for the whole series and 68% ± 11% for patients who received transplantation. Systemic (extra-CNS) and/or meningeal disease did not affect outcome. Conclusion The combination of high doses of antimetabolites, R-HDS, and ASCT is feasible and effective in patients age 18 to 70 years old with secondary CNS lymphoma, and we propose it as a new standard therapeutic option.
Collapse
Affiliation(s)
- Andrés J.M. Ferreri
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Giovanni Donadoni
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Maria Giuseppina Cabras
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Caterina Patti
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Michael Mian
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Renato Zambello
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Corrado Tarella
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Massimo Di Nicola
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Alfonso M. D'Arco
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Gianluca Doa
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Marta Bruno-Ventre
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Andrea Assanelli
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Marco Foppoli
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Giovanni Citterio
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Alessandro Fanni
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Antonino Mulè
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Federico Caligaris-Cappio
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| | - Fabio Ciceri
- Andrés J.M. Ferreri, Giovanni Donadoni, Marta Bruno-Ventre, Andrea Assanelli, Marco Foppoli, Giovanni Citterio, Federico Caligaris-Cappio, and Fabio Ciceri, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute; Massimo Di Nicola, Istituto Nazionale dei Tumori, Milano; Maria Giuseppina Cabras and Alessandro Fanni, Hospital Businco, Cagliari; Caterina Patti and Antonino Mulè, Hospital “V. Cervello,” Palermo; Michael Mian, Ospedale di Bolzano, Bolzano; Renato Zambello,
| |
Collapse
|
24
|
Raub TJ, Wishart GN, Kulanthaivel P, Staton BA, Ajamie RT, Sawada GA, Gelbert LM, Shannon HE, Sanchez-Martinez C, De Dios A. Brain Exposure of Two Selective Dual CDK4 and CDK6 Inhibitors and the Antitumor Activity of CDK4 and CDK6 Inhibition in Combination with Temozolomide in an Intracranial Glioblastoma Xenograft. Drug Metab Dispos 2015; 43:1360-71. [PMID: 26149830 DOI: 10.1124/dmd.114.062745] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/02/2015] [Indexed: 11/22/2022] Open
Abstract
Effective treatments for primary brain tumors and brain metastases represent a major unmet medical need. Targeting the CDK4/CDK6-cyclin D1-Rb-p16/ink4a pathway using a potent CDK4 and CDK6 kinase inhibitor has potential for treating primary central nervous system tumors such as glioblastoma and some peripheral tumors with high incidence of brain metastases. We compared central nervous system exposures of two orally bioavailable CDK4 and CDK6 inhibitors: abemaciclib, which is currently in advanced clinical development, and palbociclib (IBRANCE; Pfizer), which was recently approved by the U.S. Food and Drug Administration. Abemaciclib antitumor activity was assessed in subcutaneous and orthotopic glioma models alone and in combination with standard of care temozolomide (TMZ). Both inhibitors were substrates for xenobiotic efflux transporters P-glycoprotein and breast cancer resistant protein expressed at the blood-brain barrier. Brain Kp,uu values were less than 0.2 after an equimolar intravenous dose indicative of active efflux but were approximately 10-fold greater for abemaciclib than palbociclib. Kp,uu increased 2.8- and 21-fold, respectively, when similarly dosed in P-gp-deficient mice. Abemaciclib had brain area under the curve (0-24 hours) Kp,uu values of 0.03 in mice and 0.11 in rats after a 30 mg/kg p.o. dose. Orally dosed abemaciclib significantly increased survival in a rat orthotopic U87MG xenograft model compared with vehicle-treated animals, and efficacy coincided with a dose-dependent increase in unbound plasma and brain exposures in excess of the CDK4 and CDK6 Ki values. Abemaciclib increased survival time of intracranial U87MG tumor-bearing rats similar to TMZ, and the combination of abemaciclib and TMZ was additive or greater than additive. These data show that abemaciclib crosses the blood-brain barrier and confirm that both CDK4 and CDK6 inhibitors reach unbound brain levels in rodents that are expected to produce enzyme inhibition; however, abemaciclib brain levels are reached more efficiently at presumably lower doses than palbociclib and are potentially on target for a longer period of time.
Collapse
Affiliation(s)
- Thomas J Raub
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Graham N Wishart
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Palaniappan Kulanthaivel
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Brian A Staton
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Rose T Ajamie
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Geri A Sawada
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Lawrence M Gelbert
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Harlan E Shannon
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Concepcion Sanchez-Martinez
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| | - Alfonso De Dios
- Drug Disposition, Lilly Research Laboratories (T.J.R., G.N.W., P.K., B.A.S., R.T.A., G.A.S.), Division of Cancer Research (L.M.G., H.E.S.), and Discovery Chemistry Research and Technologies (A.D.D.), Eli Lilly and Company, Indianapolis, Indiana; Discovery Chemistry Research and Technologies, Eli Lilly and Company, Alcobendas, Madrid, Spain (C.S.-M.); and Covance Laboratories, Greenfield, Indiana (H.E.S.)
| |
Collapse
|
25
|
Costa DB, Shaw AT, Ou SHI, Solomon BJ, Riely GJ, Ahn MJ, Zhou C, Shreeve SM, Selaru P, Polli A, Schnell P, Wilner KD, Wiltshire R, Camidge DR, Crinò L. Clinical Experience With Crizotinib in Patients With Advanced ALK-Rearranged Non-Small-Cell Lung Cancer and Brain Metastases. J Clin Oncol 2015; 33:1881-8. [PMID: 25624436 DOI: 10.1200/jco.2014.59.0539] [Citation(s) in RCA: 495] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
PURPOSE Crizotinib is an oral kinase inhibitor approved for the treatment of ALK-rearranged non-small-cell lung cancer (NSCLC). The clinical benefits of crizotinib in patients with brain metastases have not been previously studied. PATIENTS AND METHODS Patients with advanced ALK-rearranged NSCLC enrolled onto clinical trial PROFILE 1005 or 1007 (randomly assigned to crizotinib) were included in this retrospective analysis. Patients with asymptomatic brain metastases (nontarget or target lesions) were allowed to enroll. Tumor assessments were evaluated every 6 weeks using RECIST (version 1.1). RESULTS At baseline, 31% of patients (275 of 888) had asymptomatic brain metastases; 109 had received no prior and 166 had received prior brain radiotherapy as treatment. Among patients with previously untreated asymptomatic brain metastases, the systemic disease control rate (DCR) at 12 weeks was 63% (95% CI, 54% to 72%), the intracranial DCR was 56% (95% CI, 46% to 66%), and the median intracranial time to progression (TTP) was 7 months (95% CI, 6.7 to 16.4). Among patients with previously treated brain metastases, the systemic DCR was 65% (95% CI, 57% to 72%), the intracranial DCR was 62% (95% CI, 54% to 70%), and the median intracranial TTP was 13.2 months (95% CI, 9.9 to not reached). Patients with systemic disease control were also likely to experience intracranial disease control at 12 weeks (correlation coefficient, 0.7652; P < .001). Among patients without baseline brain metastases who developed progressive disease (n = 253) after initiation of crizotinib, 20% were diagnosed with brain metastases. CONCLUSION Crizotinib was associated with systemic and intracranial disease control in patients with ALK-rearranged NSCLC who were ALK inhibitor naive and had brain metastases. However, progression of preexisting or development of new intracranial lesions while receiving therapy was a common manifestation of acquired resistance to crizotinib.
Collapse
Affiliation(s)
- Daniel B Costa
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO.
| | - Alice T Shaw
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Sai-Hong I Ou
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Benjamin J Solomon
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Gregory J Riely
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Myung-Ju Ahn
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Caicun Zhou
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - S Martin Shreeve
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Paulina Selaru
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Anna Polli
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Patrick Schnell
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Keith D Wilner
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Robin Wiltshire
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - D Ross Camidge
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| | - Lucio Crinò
- Daniel B. Costa, Beth Israel Deaconess Medical Center, Harvard Medical School; Alice T. Shaw, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Sai-Hong I. Ou, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine; S. Martin Shreeve, Paulina Selaru, and Keith D. Wilner, Pfizer Oncology, La Jolla, CA; Benjamin J. Solomon, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Gregory J. Riely, Memorial Sloan-Kettering Cancer Center; Patrick Schnell, Pfizer Oncology, New York, NY; Myung-Ju Ahn, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea; Caicun Zhou, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China; Anna Polli, Pfizer Oncology, Milan; Lucio Crinò, Perugia University Medical School, Perugia, Italy; Robin Wiltshire, Pfizer Oncology, Tadworth, United Kingdom; and D. Ross Camidge, University of Colorado Denver, Aurora, CO
| |
Collapse
|
26
|
Azeredo FJ, Dalla Costa T, Derendorf H. Role of microdialysis in pharmacokinetics and pharmacodynamics: current status and future directions. Clin Pharmacokinet 2014; 53:205-212. [PMID: 24452811 DOI: 10.1007/s40262-014-0131-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Diagnostic and therapeutic decisions in medical practice are still generally based on blood concentrations of drugs and/or biomolecules despite the knowledge that biochemical events and pharmacological effects usually take place in tissue rather than in the bloodstream. Microdialysis is a semi-invasive technique that is able to measure concentrations of the free, active drug or endogenous compounds in almost all human tissues and organs. It is currently being used to monitor brain metabolic processes and quantify tissue biomarkers, and determine transdermal drug distribution and tissue pharmacokinetics, confirming its importance as a widely used sampling technique in clinical drug monitoring and drug development as well as therapy and disease follow-up, contributing to rationalizing drug dosing regimens and influencing the clinical decision-making process.
Collapse
Affiliation(s)
| | - Teresa Dalla Costa
- Programa de Pós-Graduação em Ciências Farmacêuticas, UFRGS, Porto Alegre, Brazil
| | - Hartmut Derendorf
- Department of Pharmaceutics, College of Pharmacy, University of Florida, 1600 SW Archer Road, Gainesville, FL, 32610, USA.
| |
Collapse
|
27
|
Adkins CE, Nounou MI, Mittapalli RK, Terrell-Hall TB, Mohammad AS, Jagannathan R, Lockman PR. A novel preclinical method to quantitatively evaluate early-stage metastatic events at the murine blood-brain barrier. Cancer Prev Res (Phila) 2014; 8:68-76. [PMID: 25348853 DOI: 10.1158/1940-6207.capr-14-0225] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The observation that approximately 15% of women with disseminated breast cancer will develop symptomatic brain metastases combined with treatment guidelines discouraging single-agent chemotherapeutic strategies facilitates the desire for novel strategies aimed at outright brain metastasis prevention. Effective and robust preclinical methods to evaluate early-stage metastatic processes, brain metastases burden, and overall mean survival are lacking. Here, we develop a novel method to quantitate early metastatic events (arresting and extravasation) in addition to traditional end time-point parameters such as tumor burden and survival in an experimental mouse model of brain metastases of breast cancer. Using this method, a reduced number of viable brain-seeking metastatic cells (from 3,331 ± 263 cells/brain to 1,079 ± 495 cells/brain) were arrested in brain one week postinjection after TGFβ knockdown. Treatment with a TGFβ receptor inhibitor, galunisertib, reduced the number of arrested cells in brain to 808 ± 82 cells/brain. Furthermore, we observed a reduction in the percentage of extravasated cells (from 63% to 30%) compared with cells remaining intralumenal when TGFβ is knocked down or inhibited with galunisertib (40%). The observed reduction of extravasated metastatic cells in brain translated to smaller and fewer brain metastases and resulted in prolonged mean survival (from 36 days to 62 days). This method opens up potentially new avenues of metastases prevention research by providing critical data important to early brain metastasis of breast cancer events.
Collapse
Affiliation(s)
- Chris E Adkins
- Department of Basic Pharmaceutical Sciences, Health Sciences Center, School of Pharmacy, West Virginia University, Morgantown, West Virginia. Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Mohamed I Nounou
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas. Department of Pharmaceutics, School of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rajendar K Mittapalli
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Tori B Terrell-Hall
- Department of Basic Pharmaceutical Sciences, Health Sciences Center, School of Pharmacy, West Virginia University, Morgantown, West Virginia. Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Afroz S Mohammad
- Department of Basic Pharmaceutical Sciences, Health Sciences Center, School of Pharmacy, West Virginia University, Morgantown, West Virginia. Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas
| | - Rajaganapathi Jagannathan
- Department of Basic Pharmaceutical Sciences, Health Sciences Center, School of Pharmacy, West Virginia University, Morgantown, West Virginia
| | - Paul R Lockman
- Department of Basic Pharmaceutical Sciences, Health Sciences Center, School of Pharmacy, West Virginia University, Morgantown, West Virginia. Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, Texas.
| |
Collapse
|
28
|
Role of microdialysis in pharmacokinetics and pharmacodynamics: current status and future directions. Clin Pharmacokinet 2014. [PMID: 24452811 DOI: 10.1007/s40262–014-0131–8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
Diagnostic and therapeutic decisions in medical practice are still generally based on blood concentrations of drugs and/or biomolecules despite the knowledge that biochemical events and pharmacological effects usually take place in tissue rather than in the bloodstream. Microdialysis is a semi-invasive technique that is able to measure concentrations of the free, active drug or endogenous compounds in almost all human tissues and organs. It is currently being used to monitor brain metabolic processes and quantify tissue biomarkers, and determine transdermal drug distribution and tissue pharmacokinetics, confirming its importance as a widely used sampling technique in clinical drug monitoring and drug development as well as therapy and disease follow-up, contributing to rationalizing drug dosing regimens and influencing the clinical decision-making process.
Collapse
|
29
|
Ye L, Shi J, Wan S, Yang X, Wang Y, Zhang J, Zheng D, Liu Z. Development and validation of a liquid chromatography-tandem mass spectrometry method for topotecan determination in beagle dog plasma and its application in a bioequivalence study. Biomed Chromatogr 2013; 27:1532-1539. [PMID: 23788247 DOI: 10.1002/bmc.2956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 04/12/2013] [Accepted: 05/07/2013] [Indexed: 01/15/2023]
Abstract
Topotecan (TPT) is an important anti-cancer drug that inhibits topoisomerase I. A sensitive and robust liquid chromatography-tandem mass spectrometry (LC-MS/MS) method that potentially determines TPT in beagle dog plasma is needed for a bioequivalence study of TPT formulations. We developed and validated LC-MS/MS to evaluate TPT in beagle dog plasma in terms of specificity, linearity, precision, accuracy, stability, extraction recovery and matrix effect. Plasma samples were treated with an Ostro(TM) sorbent plate (a robust and effective tool) to eliminate phospholipids and proteins before analysis. TPT and camptothecin (internal standard) were separated on an Acquity UPLC BEH C18 column (1.7 µm, 2.1 × 50 mm) with 0.1% formic acid and methanol as the mobile phase at a flow rate of 0.25 mL/min. TPT was analyzed using positive ion electrospray ionization in multiple-reaction monitoring mode. The obtained lower limit of quantitation was 1 ng/mL (signal-to-noise ratio > 10). The standard calibration curve for TPT was linear (correlation coefficient > 0.99) at the concentration range of 1-400 ng/mL. The intra-day and inter-day precision, accuracy, stability, extraction recovery and matrix effect of TPT were within the acceptable limits. The validated method was successfully applied in a bioequivalence study of TPT in healthy beagle dogs.
Collapse
Affiliation(s)
- Ling Ye
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Thorsen F, Fite B, Mahakian LM, Seo JW, Qin S, Harrison V, Johnson S, Ingham E, Caskey C, Sundstrøm T, Meade TJ, Harter PN, Skaftnesmo KO, Ferrara KW. Multimodal imaging enables early detection and characterization of changes in tumor permeability of brain metastases. J Control Release 2013; 172:812-22. [PMID: 24161382 DOI: 10.1016/j.jconrel.2013.10.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 12/31/2022]
Abstract
Our goal was to develop strategies to quantify the accumulation of model therapeutics in small brain metastases using multimodal imaging, in order to enhance the potential for successful treatment. Human melanoma cells were injected into the left cardiac ventricle of immunodeficient mice. Bioluminescent, MR and PET imaging were applied to evaluate the limits of detection and potential for contrast agent extravasation in small brain metastases. A pharmacokinetic model was applied to estimate vascular permeability. Bioluminescent imaging after injecting d-luciferin (molecular weight (MW) 320 D) suggested that tumor cell extravasation had already occurred at week 1, which was confirmed by histology. 7T T1w MRI at week 4 was able to detect non-leaky 100 μm sized lesions and leaky tumors with diameters down to 200 μm after contrast injection at week 5. PET imaging showed that (18)F-FLT (MW 244 Da) accumulated in the brain at week 4. Gadolinium-based MRI tracers (MW 559 Da and 2.066 kDa) extravasated after 5 weeks (tumor diameter 600 μm), and the lower MW agent cleared more rapidly from the tumor (mean apparent permeabilities 2.27 × 10(-5)cm/s versus 1.12 × 10(-5)cm/s). PET imaging further demonstrated tumor permeability to (64)Cu-BSA (MW 65.55 kDa) at week 6 (tumor diameter 700 μm). In conclusion, high field T1w MRI without contrast may improve the detection limit of small brain metastases, allowing for earlier diagnosis of patients, although the smallest lesions detected with T1w MRI were permeable only to d-luciferin and the amphipathic small molecule (18)F-FLT. Different-sized MR and PET contrast agents demonstrated the gradual increase in leakiness of the blood tumor barrier during metastatic progression, which could guide clinicians in choosing tailored treatment strategies.
Collapse
Affiliation(s)
- Frits Thorsen
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Awasthi A, Lohani M, Singh MK, Singh AT, Jaggi M. Pharmacokinetic evaluation of C-3 modified 1,8-naphthyridine-3-carboxamide derivatives with potent anticancer activity: lead finding. J Enzyme Inhib Med Chem 2013; 29:710-21. [PMID: 24156741 DOI: 10.3109/14756366.2013.845817] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
To develop naphthyridine derivatives as anticancer candidates, pharmacokinetic (PK) evaluations of 10 novel derivatives of 1,4-dihydro-4-oxo-1-proparagyl-1,8-naphthyridine-3-carboxamide, with potent anticancer activity were done using in vitro ADME (absorption, distribution, metabolism, excretion) and pharmacokinetic--pharmcodynamic (PK/PD) assays. Only derivatives 5, 6, 9 and 10 showed better metabolic stability, solubility, permeability, partition coefficient and cytochrome P450 (CYP) inhibition values. PK of derivatives 5, 6, 9 and 10 in rat showed comparable PK profile for derivative 5 (C0 = 6.98 µg/mL) and 6 (C0 = 6.61 µg/mL) with no detectable plasma levels for derivatives 9 and 10 at 5.0 mg/kg i.v. dose. PK/PD assay of derivatives 5 and 6 in tumor-bearing mice (TBM) showed comparable PK but tumor plasma index (TPI) of derivative 6 (4.02) was better than derivative 5 (2.50), suggesting better tumor uptake of derivative 6. Derivative 6, as lead compound, showed highest tumor growth inhibition (TGI) value of 33.6% in human ovary cancer xenograft model.
Collapse
Affiliation(s)
- Anshumali Awasthi
- Department of Drug Metabolism and Pharmacokinetics (DMPK), Dabur Research Foundation, Ghaziabad , Uttar Pradesh , India and
| | | | | | | | | |
Collapse
|
32
|
Martínez-Aranda A, Hernández V, Picón C, Modolell I, Sierra A. Development of a preclinical therapeutic model of human brain metastasis with chemoradiotherapy. Int J Mol Sci 2013; 14:8306-27. [PMID: 23591844 PMCID: PMC3645744 DOI: 10.3390/ijms14048306] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/16/2013] [Accepted: 03/26/2013] [Indexed: 01/22/2023] Open
Abstract
Currently, survival of breast cancer patients with brain metastasis ranges from 2 to 16 months. In experimental brain metastasis studies, only 10% of lesions with the highest permeability exhibited cytotoxic responses to paclitaxel or doxorubicin. Therefore, radiation is the most frequently used treatment, and sensitizing agents, which synergize with radiation, can improve the efficacy of the therapy. In this study we used 435-Br1 cells containing the fluorescent protein (eGFP) gene and the photinus luciferase (PLuc) gene to develop a new brain metastatic cell model in mice through five in vivo/in vitro rounds. BR-eGFP-CMV/Luc-V5 brain metastatic cells induce parenchymal brain metastasis within 60.8 ± 13.8 days of intracarotid injection in all mice. We used this model to standardize a preclinical chemoradiotherapy protocol comprising three 5.5 Gy fractions delivered on consecutive days (overall dose of 16.5 Gy) which improved survival with regard to controls (60.29 ± 8.65 vs. 47.20 ± 11.14). Moreover, the combination of radiotherapy with temozolomide, 60 mg/Kg/day orally for five consecutive days doubled survival time of the mice 121.56 ± 52.53 days (Kaplan-Meier Curve, p < 0.001). This new preclinical chemoradiotherapy protocol proved useful for the study of radiation response/resistance in brain metastasis, either alone or in combination with new sensitizing agents.
Collapse
Affiliation(s)
- Antonio Martínez-Aranda
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L’ Hospitalet de Llobregat, Barcelona 08907, Spain; E-Mails: (A.M.-A.); (V.H.)
- Autonoma University of Barcelona (UAB), Faculty of Biosciences, Campus Bellaterra, Building C, Cerdanyola del Vallés, Barcelona 08193, Spain
| | - Vanessa Hernández
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L’ Hospitalet de Llobregat, Barcelona 08907, Spain; E-Mails: (A.M.-A.); (V.H.)
| | - Cristina Picón
- Medical Physics Service, Oncology Catalan Institut, Duran I Reynals Hospital, L’Hospitalet de Llobregat, Barcelona 08907, Spain; E-Mails: (C.P.); (I.M.)
| | - Ignasi Modolell
- Medical Physics Service, Oncology Catalan Institut, Duran I Reynals Hospital, L’Hospitalet de Llobregat, Barcelona 08907, Spain; E-Mails: (C.P.); (I.M.)
| | - Angels Sierra
- Biological Clues of the Invasive and Metastatic Phenotype Group, Bellvitge Biomedical Research Institute (IDIBELL), L’ Hospitalet de Llobregat, Barcelona 08907, Spain; E-Mails: (A.M.-A.); (V.H.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-93-260-7429, Fax: +34-93-260-7426
| |
Collapse
|
33
|
Li L, Agarwal S, Elmquist WF. Brain efflux index to investigate the influence of active efflux on brain distribution of pemetrexed and methotrexate. Drug Metab Dispos 2013; 41:659-67. [PMID: 23297298 PMCID: PMC3920090 DOI: 10.1124/dmd.112.049254] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 01/04/2013] [Indexed: 01/07/2023] Open
Abstract
Antifolates, in particular methotrexate (MTX), have been widely used in the treatment of primary and secondary tumors of the central nervous system (CNS). Pemetrexed (PMX) is a novel antifolate that also exhibits potent antitumor activity against CNS malignancies. Studies have shown that brain distribution of both antifolates is significantly restricted, possible due to active efflux transport at the blood-brain barrier (BBB). This study characterizes the brain-to-blood transport of PMX and MTX and examines the role of several efflux transporters in brain distribution of the antifolates by use of the intracerebral microinjection technique (brain efflux index). The results from this study show that both PMX and MTX undergo saturable efflux transport across the BBB, with elimination half-lives of approximately 39 minutes and 29 minutes, respectively. Of the various efflux transporters this study investigated, multidrug resistance-associated protein 2 (Mrp2) does not play an important role in the brain distribution of the two antifolate drugs. Interestingly, breast-cancer resistance protein (Bcrp) makes a significant contribution to the brain elimination of MTX but not PMX. In addition, the brain-to-blood transport of both antifolates was inhibited by probenecid and benzylpenicillin, suggesting the involvement of organic anion transporters in the efflux of these compounds from the brain, with organic anion transporter 3 (Oat3) being a possibility. Our results suggest that one of the underlying mechanisms behind the limited brain distribution of PMX and MTX is active efflux transport processes at the BBB, including a benzylpenicillin-sensitive transport system and/or the active transporter Bcrp.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
34
|
Lv H, Zhang X, Sharma J, Reddy MVR, Reddy EP, Gallo JM. Integrated pharmacokinetic-driven approach to screen candidate anticancer drugs for brain tumor chemotherapy. AAPS JOURNAL 2012. [PMID: 23180160 DOI: 10.1208/s12248-012-9428-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The goal of the study was to develop an effective screening strategy to select new agents for brain tumor chemotherapy from a series of low molecular weight anticancer agents [ON123x] by the combined use of in silico, in vitro cytotoxicity, and in vitro ADME profiling studies. The results of these studies were cast into a pipeline of tier 1 and tier 2 procedures that resulted in the identification of ON123300 as the lead compound. Of the 154 ON123xx compounds, 13 met tier 1 screening criteria based on physicochemical properties [i.e., MW < 450 Da, predicted log P between 2 and 3.5] and in vitro glioma cell cytotoxicity [i.e., IC50 < 10 μM] and were further tested in tier 2 assays. The tier 2 profiling studies consisted of metabolic stability, MDCK-MDR1 cell permeability and plasma and brain protein binding that were combined to globally assess whether favorable pharmacokinetic properties and brain penetration could be achieved in vivo. In vivo cassette dosing studies were conducted in mice for 12 compounds that permitted examination of in vitro/in vivo relationships that confirmed the suitability of the in vitro assays. A parameter derived from the in vitro assays accurately predicted the extent of drug accumulation in the brain based on the area under the drug concentration-time curve in brain measured in the cassette dosing study (r (2) = 0.920). Overall, the current studies demonstrated the value of an integrated pharmacokinetic-driven approach to identify potentially efficacious agents for brain tumor chemotherapy.
Collapse
Affiliation(s)
- Hua Lv
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
35
|
Martins SJ, Azevedo CRASD, Chinen LTD, Cruz MRS, Peterlevitz MA, Gimenes DL. Meningeal carcinomatosis in solid tumors. ARQUIVOS DE NEURO-PSIQUIATRIA 2012; 69:973-80. [PMID: 22297890 DOI: 10.1590/s0004-282x2011000700024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 07/01/2011] [Indexed: 02/06/2023]
Abstract
The involvement of the leptomeninges by metastatic tumors can be observed in solid tumors, in which case it is termed meningeal carcinomatosis (MC), and in lymphoproliferative malignant disease. It is more common in breast and lung cancer, as well as melanoma, with adenocarcinoma being the most frequent histological type. MC is usually a late event, with disseminated and progressive disease already present and, it is characterized by multifocal neurological signs and symptoms. Diagnosis is based on the evaluation of clinical presentation, cerebrospinal fluid and neuroimaging studies. The better systemic disease control is observed with new therapeutic agents, and the development of neuroimaging methods is responsible for the increasing incidence of such metastatic evolution. Intrathecal chemotherapy is generally the treatment of choice, although frequently palliative. Prognosis is guarded, although a higher performance status may indicate a subgroup of patients with a more favorable outcome.
Collapse
|
36
|
Joerger M, Huitema ADR, Illerhaus G, Ferreri AJM. Rational administration schedule for high-dose methotrexate in patients with primary central nervous system lymphoma. Leuk Lymphoma 2012; 53:1867-75. [PMID: 22530664 DOI: 10.3109/10428194.2012.676177] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Methotrexate (MTX) at a dose of ≥1 g/m(2) remains the most efficient treatment against primary central nervous system lymphoma (PCNSL), and is the most widely used drug in prospective clinical trials. MTX is a folate analog that inhibits dihydrofolate reductase, thereby blocking de novo purine synthesis. MTX as well as 7-hydroxy-MTX, its main metabolite in serum, are both eliminated by the kidneys. The elimination of MTX is prolonged in patients with renal impairment and third-space fluid collections, and in patients receiving concurrent non-steroidal antirheumatic drugs, benzimidazoles and sulfonamides, among others. Main adverse events with high-dose MTX include severe myelosuppression, renal dysfunction and stomatitis. Supportive measures such as rigorous hydration, urine alkalinization and careful drug monitoring with supplemental leucovorin rescue are crucial to avoid significant toxicity. Strategies to optimize clinical efficacy of high-dose MTX in patients with PCNSL include administration of 3 h instead of longer infusions, potentially supplemented with an additional intravenous MTX bolus, and maintaining MTX dose intensity over the course of four treatment cycles. Some pharmacological studies suggest that achieving an MTX area under the plasma concentration-time curve (AUC(MTX)) of between 1000 and 1100 μmol.h/L may improve clinical outcome, but clinical data are not conclusive at present. In this review, we analyze the impact of patient, lymphoma and pharmacokinetic variables on the antitumor activity of high-dose MTX in patients with PCNSL, summarize recommendations for daily clinical practice and give some suggestions for future trials.
Collapse
Affiliation(s)
- M Joerger
- Department of Oncology and Hematology, Cantonal Hospital, St. Gallen, Switzerland.
| | | | | | | |
Collapse
|
37
|
Doloff JC, Waxman DJ. VEGF receptor inhibitors block the ability of metronomically dosed cyclophosphamide to activate innate immunity-induced tumor regression. Cancer Res 2012; 72:1103-15. [PMID: 22237627 DOI: 10.1158/0008-5472.can-11-3380] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In metronomic chemotherapy, frequent drug administration at lower than maximally tolerated doses can improve activity while reducing the dose-limiting toxicity of conventional dosing schedules. Although the antitumor activity produced by metronomic chemotherapy is attributed widely to antiangiogenesis, the significance of this mechanism remains somewhat unclear. In this study, we show that a 6-day repeating metronomic schedule of cyclophosphamide administration activates a potent antitumor immune response associated with brain tumor recruitment of natural killer (NK) cells, macrophages, and dendritic cells that leads to marked tumor regression. Tumor regression was blocked in nonobese diabetic/severe combined immunodeficient (NOD/SCID-γ) mice, which are deficient or dysfunctional in all these immune cell types. Furthermore, regression was blunted by NK cell depletion in immunocompetent syngeneic mice or in perforin-deficient mice, which are compromised for NK, NKT, and T-cell cytolytic functions. Unexpectedly, we found that VEGF receptor inhibitors blocked both innate immune cell recruitment and the associated tumor regression response. Cyclophosphamide administered at a maximum tolerated dose activated a transient, weak innate immune response, arguing that persistent drug-induced cytotoxic damage or associated cytokine and chemokine responses are required for effective innate immunity-based tumor regression. Together, our results reveal an innate immunity-based mechanism of tumor regression that can be activated by a traditional cytotoxic chemotherapy administered on a metronomic schedule. These findings suggest the need to carefully evaluate the clinical effects of combination chemotherapies that incorporate antiangiogenesis drugs targeting VEGF receptor.
Collapse
Affiliation(s)
- Joshua C Doloff
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
38
|
Brain Metastases as the Primary Site of Relapse in Two Randomized Phase III Pemetrexed Trials in Advanced Non–Small-Cell Lung Cancer. Clin Lung Cancer 2012; 13:24-30. [DOI: 10.1016/j.cllc.2011.05.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 05/26/2011] [Accepted: 05/31/2011] [Indexed: 11/17/2022]
|
39
|
Sathornsumetee S, Rich JN. Molecularly targeted therapy in neuro-oncology. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:255-78. [PMID: 22230448 DOI: 10.1016/b978-0-444-52138-5.00018-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
40
|
Alimohamed N, Daly A, Owen C, Duggan P, Stewart DA. Upfront thiotepa, busulfan, cyclophosphamide, and autologous stem cell transplantation for primary CNS lymphoma: a single centre experience. Leuk Lymphoma 2011; 53:862-7. [PMID: 22023529 DOI: 10.3109/10428194.2011.633250] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Treatment of primary central nervous system lymphoma (PCNSL) with high-dose methotrexate-based chemotherapy and whole-brain radiotherapy (WBRT) is associated with high rates of relapse and severe neurotoxicity. In an attempt to improve upon these poor results, we treated 21 patients with PCNSL aged 34-69 years (median 56) with high-dose thiotepa, busulfan, cyclophosphamide (TBC) and autologous stem cell transplant (ASCT) as part of front-line therapy, without WBRT. Patient characteristics included: Karnofsky performance status (KPS) <70% (n = 17), age >60 years (n = 8), deep brain involvement (n = 16). Treatment-induced neurotoxicity was not observed in any of these patients. Currently, 11 of 21 patients (52%) are alive and progression-free at a median follow-up of 60 (7-125) months post-ASCT. Causes of death included progressive PCNSL (n = 4), progressive systemic lymphoma (n = 1), early treatment-related mortality (TRM, n = 3) and two late deaths from pneumonia 3 years post-ASCT. All patients who died of TRM were over 60 years of age and had poor performance status. In conclusion, TBC/ASCT offers potential long-term progression-free survival without neurotoxicity when used as part of upfront therapy for PCNSL. However, efforts to reduce TRM through improved patient selection and possibly through decreased intensity of the TBC regimen for older or less fit patients are recommended.
Collapse
Affiliation(s)
- Nimira Alimohamed
- Department of Oncology and Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
41
|
Haque S, Md S, Alam MI, Sahni JK, Ali J, Baboota S. Nanostructure-based drug delivery systems for brain targeting. Drug Dev Ind Pharm 2011; 38:387-411. [PMID: 21954902 DOI: 10.3109/03639045.2011.608191] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CONTEXT It is well-known fact that blood brain barrier (BBB) hinders the penetrance and access of many pharmacotherapeutic agents to central nervous system (CNS). Many diseases of the CNS remain undertreated and the inability to treat most CNS disorders is not due to the lack of effective CNS drug discovery, rather, it is due to the ineffective CNS delivery. Therefore, a number of nanostructured drug delivery carriers have been developed and explored over the past couple of years to transport the drugs to brain. OBJECTIVE The present review will give comprehensive details of extensive research being done in field of nanostructured carriers to transport the drugs through the BBB in a safe and effective manner. METHODS The method includes both the polymeric- and lipid-based nanocarriers with emphasis on their utility, methodology, advantages, and the drugs which have been worked on using a particular approach to provide a noninvasive method to improve the drug transport through BBB. RESULTS Polymeric- and lipid-based nanocarriers enter brain capillaries before reaching the surface of the brain microvascular endothelial cells without the disruption of BBB. These systems are further modified with specific ligands vectors and pegylation aiming to target and enhance their binding with surface receptors of the specific tissues inside brain and increase long circulatory time which favors interaction and penetration into brain endothelial cells. CONCLUSION This review would give an insight to the researchers working on neurodegenerative and non-neurodegenerative diseases of the CNS including brain tumor.
Collapse
|
42
|
Costa DB, Kobayashi S, Pandya SS, Yeo WL, Shen Z, Tan W, Wilner KD. CSF concentration of the anaplastic lymphoma kinase inhibitor crizotinib. J Clin Oncol 2011; 29:e443-5. [PMID: 21422405 DOI: 10.1200/jco.2010.34.1313] [Citation(s) in RCA: 498] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Daniel B Costa
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Pitz MW, Desai A, Grossman SA, Blakeley JO. Tissue concentration of systemically administered antineoplastic agents in human brain tumors. J Neurooncol 2011; 104:629-38. [PMID: 21400119 DOI: 10.1007/s11060-011-0564-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 03/03/2011] [Indexed: 12/24/2022]
Abstract
The blood-brain-barrier (BBB) limits the penetration of many systemic antineoplastic therapies. Consequently, many agents may be used in clinical studies and clinical practice though they may not achieve therapeutic levels within the tumor. We sought to compile the currently available human data on antineoplastic drug concentrations in brain and tumor tissue according to BBB status. A review of the literature was conducted for human studies providing concentrations of antineoplastic agents in blood and metastatic brain tumors or high-grade gliomas. Studies were considered optimal if they reported simultaneous tissue and blood concentration, multiple sampling times and locations, MRI localization, BBB status at sampling site, tumor histology, and individual subject data. Twenty-Four studies of 19 compounds were included. These examined 18 agents in contrast-enhancing regions of high-grade gliomas, with optimal data for 2. For metastatic brain tumors, adequate data was found for 9 agents. Considerable heterogeneity was found in the measurement value, tumor type, measurement timing, and sampling location within and among studies, limiting the applicability of the results. Tissue to blood ratios ranged from 0.054 for carboplatin to 34 for mitoxantrone in high-grade gliomas, and were lowest for temozolomide (0.118) and etoposide (0.116), and highest for mitoxantrone (32.02) in metastatic tumors. The available data examining the concentration of antineoplastic agents in brain and tumor tissue is sparse and limited by considerable heterogeneity. More studies with careful quantification of antineoplastic agents in brain and tumor tissue is required for the rational development of therapeutic regimens.
Collapse
Affiliation(s)
- Marshall W Pitz
- University of Manitoba, 675 McDermot Avenue, Winnipeg, MB, R3E 0V9, Canada.
| | | | | | | |
Collapse
|
44
|
Minturn JE, Janss AJ, Fisher PG, Allen JC, Patti R, Phillips PC, Belasco JB. A phase II study of metronomic oral topotecan for recurrent childhood brain tumors. Pediatr Blood Cancer 2011; 56:39-44. [PMID: 21108437 DOI: 10.1002/pbc.22690] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND The prognosis for recurrent or refractory brain tumors in children is poor with conventional therapies. Topotecan is a topoisomerase I inhibitor with good central nervous system (CNS) penetration following oral administration. Increased efficacy of topotecan has been demonstrated with prolonged low-dose daily treatment in pre-clinical models. To investigate further this drug delivered orally in pediatric CNS malignancies, a phase II study in children with recurrent or refractory brain tumors was performed. PROCEDURE Patients ≤ 21 years of age at diagnosis with a recurrent, progressive, or refractory primary CNS malignancy and measurable disease, were eligible. Patients enrolled into four strata: ependymoma (N = 4), high-grade glioma (HGG) (N = 6), brainstem glioma (BSG) (N = 13), and primitive neuroectodermal tumor (PNET) (N = 8). Oral topotecan was administered once daily at a dose of 0.8 mg/m(2)/day for 21 consecutive days repeated every 28 days. Response and toxicity profiles were evaluated. RESULTS Twenty-six patients were evaluable (median age 9.2 years; 10 males). Two objective responses were observed in PNET patients with disseminated tumor at study entry. These two patients remain alive and in remission 7 and 9.5 years off study. Four other patients (two BSG, one PNET, and one HGG) had stable disease (median 4.6 months). The most common toxicities were hematologic. CONCLUSIONS Daily oral topotecan at a dose of 0.8 mg/m(2)/day can be safely administered to children with recurrent or refractory brain tumors. This regimen identified activity in recurrent PNET. The prolonged progression free survival (PFS) in two PNET patients justifies consideration of this regimen in more advanced clinical trials.
Collapse
Affiliation(s)
- Jane E Minturn
- Division of Oncology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Cruz-Muñoz W, Kerbel RS. Preclinical approaches to study the biology and treatment of brain metastases. Semin Cancer Biol 2010; 21:123-30. [PMID: 21147227 DOI: 10.1016/j.semcancer.2010.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 12/02/2010] [Indexed: 12/22/2022]
Abstract
Metastatic spread to the central nervous system (CNS) is a common and devastating manifestation of major cancer types. Its incidence is associated with poor prognosis manifested by neurological deterioration leading to diminished quality of life and an extremely short median survival. CNS metastasis is becoming an increasingly important clinical problem. This is especially the case for certain types of cancers for which effective treatments of visceral disease are available. As a result of the present limitations in treating CNS metastases, this manifestation of tumor progression remains an unmet clinical need. Despite its significance, our general understanding of the mechanisms that regulate the brain-metastatic phenotype is currently meager. Both the analysis of mechanistic aspects of brain metastasis and the development of effective treatments necessitate the use of appropriate in vivo models that recapitulate the interaction of the tumor cells with the microenvironment of the brain. Here we review the available preclinical models of CNS metastasis and their use as tools to advance knowledge of the biology of the disease (with the aim of identifying relevant molecular determinants, prognostic biomarkers, and therapeutic targets) as well as examining effective approaches for treatment.
Collapse
Affiliation(s)
- William Cruz-Muñoz
- Sunnybrook Health Sciences Centre, Molecular and Cellular Biology Research, S-217, 2075 Bayview Ave., Toronto, Ontario M4N 3M5, Canada
| | | |
Collapse
|
46
|
Drug delivery to the brain: considerations of genetic polymorphisms of blood-brain barrier transporters and imaging technologies. Ther Deliv 2010; 1:563-70. [PMID: 22833967 DOI: 10.4155/tde.10.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PubMed search and literature reviews summarized our latest advances regarding the impact of genetic polymorphisms of blood-brain barrier transporters on, and the application of imaging technologies to evaluate and enhance drug delivery to the brain. Although there are reports relating transporter polymorphisms to pharmacokinetics or adverse events, few studies have extended such efforts to the brain. Conceivably, simultaneous nonfunctional expressions of more than one key efflux transporters could result in devastating clinical outcomes if the dose of their substrate drug is not adjusted for the subpopulation with such a phenotype. Imaging technologies have been used to elucidate the kinetic or functional activities of blood-brain barrier transporters, with the majority focusing on P-glycoprotein. Imaging technologies have been used to discover drugs for treating brain disorders, enable targeted delivery in combination with convection-enhanced delivery, assess the therapeutic effect of a treatment protocol and assess drug penetration into brain tumors. Research is limited, however, in using imaging technologies to link the genotype or phenotype of a transporter to the uptake of drug into the brain. No imaging studies have provided clear evidence of transporter polymorphisms discriminating the distribution of drug in the brain. Research efforts are needed to use imaging technologies to explore the correlation between pharmacogenetics and individual clinical outcomes, especially for the phenotype of more than one nonfunctional transporters.
Collapse
|
47
|
Lockman PR, Mittapalli RK, Taskar KS, Rudraraju V, Gril B, Bohn KA, Adkins CE, Roberts A, Thorsheim HR, Gaasch JA, Huang S, Palmieri D, Steeg PS, Smith QR. Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clin Cancer Res 2010; 16:5664-78. [PMID: 20829328 DOI: 10.1158/1078-0432.ccr-10-1564] [Citation(s) in RCA: 522] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Brain metastases of breast cancer appear to be increasing in incidence, confer significant morbidity, and threaten to compromise gains made in systemic chemotherapy. The blood-tumor barrier (BTB) is compromised in many brain metastases; however, the extent to which this influences chemotherapeutic delivery and efficacy is unknown. Herein, we answer this question by measuring BTB passive integrity, chemotherapeutic drug uptake, and anticancer efficacy in vivo in two breast cancer models that metastasize preferentially to brain. EXPERIMENTAL DESIGN Experimental brain metastasis drug uptake and BTB permeability were simultaneously measured using novel fluorescent and phosphorescent imaging techniques in immune-compromised mice. Drug-induced apoptosis and vascular characteristics were assessed using immunofluorescent microscopy. RESULTS Analysis of over 2,000 brain metastases from two models (human 231-BR-Her2 and murine 4T1-BR5) showed partial BTB permeability compromise in greater than 89% of lesions, varying in magnitude within and between metastases. Brain metastasis uptake of ¹⁴C-paclitaxel and ¹⁴C-doxorubicin was generally greater than normal brain but less than 15% of that of other tissues or peripheral metastases, and only reached cytotoxic concentrations in a small subset (∼10%) of the most permeable metastases. Neither drug significantly decreased the experimental brain metastatic ability of 231-BR-Her2 tumor cells. BTB permeability was associated with vascular remodeling and correlated with overexpression of the pericyte protein desmin. CONCLUSIONS This work shows that the BTB remains a significant impediment to standard chemotherapeutic delivery and efficacy in experimental brain metastases of breast cancer. New brain permeable drugs will be needed. Evidence is presented for vascular remodeling in BTB permeability alterations.
Collapse
Affiliation(s)
- Paul R Lockman
- Department of Pharmaceutical Sciences, Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Vecht CJ, Wilms EB. Seizures in low- and high-grade gliomas: current management and future outlook. Expert Rev Anticancer Ther 2010; 10:663-669. [DOI: 10.1586/era.10.48] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
49
|
Gilheeney SW, Khakoo Y, Souweidane M, Wolden S, Boulad F, Dunkel IJ. Thiotepa/topotecan/carboplatin with autologous stem cell rescue in recurrent/refractory/poor prognosis pediatric malignancies of the central nervous system. Pediatr Blood Cancer 2010; 54:591-5. [PMID: 19998470 DOI: 10.1002/pbc.22347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Thiotepa and carboplatin are known to be active in central nervous system tumors. Topotecan potentiates the anti-cancer effects of alkylators and crosses the blood-brain barrier. We present ten patients with recurrent or progressive central nervous system malignancies treated on a myeloablative regimen using these drugs. METHODS Treatment included: Thiotepa 300 mg/m(2) on days -8, -7, and -6; topotecan 2 mg/m(2) on days -8, -7, -6, -5, and -4; and carboplatin approximately 500 mg/m(2) (Calvert formula-area under the curve = 7) on days -5, -4, and -3. Stem cell rescue was on day 0. RESULTS Age at study entry ranged from 2.5 to 20 years old (median age 8.7 years). Five had medulloblastoma (MB), four had high grade glioma (HGG), and one had trilateral retinoblastoma/pineoblastoma (tRB/PB). Prior treatment for all patients included surgery and chemotherapy (1-7 regimens, median 2). Nine patients received radiotherapy; one patient did not receive radiotherapy pre-study. Three patients had residual disease at the time of transplant. There were two toxic deaths. Four patients are event-free survivors at a median of 6 years (range 2.8-7.6 years) after treatment including 2/5 MB patients, 1/4 HGG patients, and the tRB/PB patient. Four of the seven patients with no evidence of disease/minimal residual disease status at the time of stem cell rescue are long-term survivors versus 1/3 with measurable disease. CONCLUSION Thiotepa/topotecan/carboplatin may help consolidate remission of poor prognosis pediatric central nervous system tumors. Diagnosis and extent of disease prior to stem cell rescue may have an impact on outcome.
Collapse
Affiliation(s)
- Stephen W Gilheeney
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Cloonan SM, Keating JJ, Butler SG, Knox AJ, Jørgensen AM, Peters GH, Rai D, Corrigan D, Lloyd DG, Williams DC, Meegan MJ. Synthesis and serotonin transporter activity of sulphur-substituted α-alkyl phenethylamines as a new class of anticancer agents. Eur J Med Chem 2009; 44:4862-88. [DOI: 10.1016/j.ejmech.2009.07.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Revised: 07/08/2009] [Accepted: 07/30/2009] [Indexed: 11/25/2022]
|