1
|
Valerius AR, Webb LM, Thomsen A, Lehrer EJ, Breen WG, Campian JL, Riviere-Cazaux C, Burns TC, Sener U. Review of Novel Surgical, Radiation, and Systemic Therapies and Clinical Trials in Glioblastoma. Int J Mol Sci 2024; 25:10570. [PMID: 39408897 PMCID: PMC11477105 DOI: 10.3390/ijms251910570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. Despite an established standard of care including surgical resection, radiation therapy, and chemotherapy, GBM unfortunately is associated with a dismal prognosis. Therefore, researchers are extensively evaluating avenues to expand GBM therapy and improve outcomes in patients with GBM. In this review, we provide a broad overview of novel GBM therapies that have recently completed or are actively undergoing study in clinical trials. These therapies expand across medical, surgical, and radiation clinical trials. We additionally review methods for improving clinical trial design in GBM.
Collapse
Affiliation(s)
| | - Lauren M. Webb
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
| | - Anna Thomsen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
| | - Eric J. Lehrer
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - William G. Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jian L. Campian
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Terry C. Burns
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
2
|
Schulz JA, Hartz AMS, Bauer B. ABCB1 and ABCG2 Regulation at the Blood-Brain Barrier: Potential New Targets to Improve Brain Drug Delivery. Pharmacol Rev 2023; 75:815-853. [PMID: 36973040 PMCID: PMC10441638 DOI: 10.1124/pharmrev.120.000025] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
The drug efflux transporters ABCB1 and ABCG2 at the blood-brain barrier limit the delivery of drugs into the brain. Strategies to overcome ABCB1/ABCG2 have been largely unsuccessful, which poses a tremendous clinical problem to successfully treat central nervous system (CNS) diseases. Understanding basic transporter biology, including intracellular regulation mechanisms that control these transporters, is critical to solving this clinical problem.In this comprehensive review, we summarize current knowledge on signaling pathways that regulate ABCB1/ABCG2 at the blood-brain barrier. In Section I, we give a historical overview on blood-brain barrier research and introduce the role that ABCB1 and ABCG2 play in this context. In Section II, we summarize the most important strategies that have been tested to overcome the ABCB1/ABCG2 efflux system at the blood-brain barrier. In Section III, the main component of this review, we provide detailed information on the signaling pathways that have been identified to control ABCB1/ABCG2 at the blood-brain barrier and their potential clinical relevance. This is followed by Section IV, where we explain the clinical implications of ABCB1/ABCG2 regulation in the context of CNS disease. Lastly, in Section V, we conclude by highlighting examples of how transporter regulation could be targeted for therapeutic purposes in the clinic. SIGNIFICANCE STATEMENT: The ABCB1/ABCG2 drug efflux system at the blood-brain barrier poses a significant problem to successful drug delivery to the brain. The article reviews signaling pathways that regulate blood-brain barrier ABCB1/ABCG2 and could potentially be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Anika M S Hartz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| |
Collapse
|
3
|
Di Nunno V, Gatto L, Tosoni A, Bartolini S, Franceschi E. Implications of BRAF V600E mutation in gliomas: Molecular considerations, prognostic value and treatment evolution. Front Oncol 2023; 12:1067252. [PMID: 36686797 PMCID: PMC9846085 DOI: 10.3389/fonc.2022.1067252] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Gliomas are molecularly heterogeneous brain tumors responsible for the most years of life lost by any cancer. High-grade gliomas have a poor prognosis and despite multimodal treatment including surgery, radiotherapy, and chemotherapy, exhibit a high recurrence rate. There is a need for new therapeutic approaches based on precision medicine informed by biomarker assessment and BRAF, a key regulator of MAPK signaling pathway, influencing cell differentiation, proliferation, migration and pro-tumorigenic activity, is emerging as a promising molecular target. V600E, is the most frequent BRAF alteration in gliomas, especially in pediatric low-grade astrocytomas, pleomorphic xanthoastrocytoma, papillary craniopharyngioma, epithelioid glioblastoma and ganglioglioma. The possible application of BRAF-targeted therapy in gliomas is continuously growing and there is preliminary evidence of prolonged disease control obtained by BRAF inhibitors in tumors harboring BRAF V600E mutation. The possibility of introducing targeted therapies into the treatment algorithm represents a paradigm shift for patients with BRAF V600E mutant recurrent high-grade and low-grade glioma and BRAF routine testing should be considered in clinical practice. The focus of this review is to summarize the molecular landscape of BRAF across glioma subtypes and the novel therapeutic strategies for BRAF V600E mutated tumors.
Collapse
Affiliation(s)
| | - Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy,*Correspondence: Lidia Gatto,
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
4
|
Ng TSC, Hu H, Kronister S, Lee C, Li R, Gerosa L, Stopka SA, Burgenske DM, Khurana I, Regan MS, Vallabhaneni S, Putta N, Scott E, Matvey D, Giobbie-Hurder A, Kohler RH, Sarkaria JN, Parangi S, Sorger PK, Agar NYR, Jacene HA, Sullivan RJ, Buchbinder E, Mikula H, Weissleder R, Miller MA. Overcoming differential tumor penetration of BRAF inhibitors using computationally guided combination therapy. SCIENCE ADVANCES 2022; 8:eabl6339. [PMID: 35486732 PMCID: PMC9054019 DOI: 10.1126/sciadv.abl6339] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/15/2022] [Indexed: 05/02/2023]
Abstract
BRAF-targeted kinase inhibitors (KIs) are used to treat malignancies including BRAF-mutant non-small cell lung cancer, colorectal cancer, anaplastic thyroid cancer, and, most prominently, melanoma. However, KI selection criteria in patients remain unclear, as are pharmacokinetic/pharmacodynamic (PK/PD) mechanisms that may limit context-dependent efficacy and differentiate related drugs. To address this issue, we imaged mouse models of BRAF-mutant cancers, fluorescent KI tracers, and unlabeled drug to calibrate in silico spatial PK/PD models. Results indicated that drug lipophilicity, plasma clearance, faster target dissociation, and, in particular, high albumin binding could limit dabrafenib action in visceral metastases compared to other KIs. This correlated with retrospective clinical observations. Computational modeling identified a timed strategy for combining dabrafenib and encorafenib to better sustain BRAF inhibition, which showed enhanced efficacy in mice. This study thus offers principles of spatial drug action that may help guide drug development, KI selection, and combination.
Collapse
Affiliation(s)
- Thomas S. C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiyu Hu
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Stefan Kronister
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Chanseo Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Luca Gerosa
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sylwia A. Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Ishaan Khurana
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Michael S. Regan
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sreeram Vallabhaneni
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Niharika Putta
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Ella Scott
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Dylan Matvey
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter K. Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nathalie Y. R. Agar
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Heather A. Jacene
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Hannes Mikula
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Institute of Applied Synthetic Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Borsari C, De Pascale M, Wymann MP. Chemical and Structural Strategies to Selectively Target mTOR Kinase. ChemMedChem 2021; 16:2744-2759. [PMID: 34114360 PMCID: PMC8518124 DOI: 10.1002/cmdc.202100332] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Indexed: 11/08/2022]
Abstract
Dysregulation of the mechanistic target of rapamycin (mTOR) pathway is implicated in cancer and neurological disorder, which identifies mTOR inhibition as promising strategy for the treatment of a variety of human disorders. First-generation mTOR inhibitors include rapamycin and its analogues (rapalogs) which act as allosteric inhibitors of TORC1. Structurally unrelated, ATP-competitive inhibitors that directly target the mTOR catalytic site inhibit both TORC1 and TORC2. Here, we review investigations of chemical scaffolds explored for the development of highly selective ATP-competitive mTOR kinase inhibitors (TORKi). Extensive medicinal chemistry campaigns allowed to overcome challenges related to structural similarity between mTOR and the phosphoinositide 3-kinase (PI3K) family. A broad region of chemical space is covered by TORKi. Here, the investigation of chemical substitutions and physicochemical properties has shed light on the compounds' ability to cross the blood brain barrier (BBB). This work provides insights supporting the optimization of TORKi for the treatment of cancer and central nervous system disorders.
Collapse
Affiliation(s)
- Chiara Borsari
- Department of BiomedicineUniversity of BaselMattenstrasse 284058BaselSwitzerland
| | - Martina De Pascale
- Department of BiomedicineUniversity of BaselMattenstrasse 284058BaselSwitzerland
| | - Matthias P. Wymann
- Department of BiomedicineUniversity of BaselMattenstrasse 284058BaselSwitzerland
| |
Collapse
|
6
|
He C, Xu K, Zhu X, Dunphy PS, Gudenas B, Lin W, Twarog N, Hover LD, Kwon CH, Kasper LH, Zhang J, Li X, Dalton J, Jonchere B, Mercer KS, Currier DG, Caufield W, Wang Y, Xie J, Broniscer A, Wetmore C, Upadhyaya SA, Qaddoumi I, Klimo P, Boop F, Gajjar A, Zhang J, Orr BA, Robinson GW, Monje M, Freeman Iii BB, Roussel MF, Northcott PA, Chen T, Rankovic Z, Wu G, Chiang J, Tinkle CL, Shelat AA, Baker SJ. Patient-derived models recapitulate heterogeneity of molecular signatures and drug response in pediatric high-grade glioma. Nat Commun 2021; 12:4089. [PMID: 34215733 PMCID: PMC8253809 DOI: 10.1038/s41467-021-24168-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 05/25/2021] [Indexed: 01/02/2023] Open
Abstract
Pediatric high-grade glioma (pHGG) is a major contributor to cancer-related death in children. In vitro and in vivo disease models reflecting the intimate connection between developmental context and pathogenesis of pHGG are essential to advance understanding and identify therapeutic vulnerabilities. Here we report establishment of 21 patient-derived pHGG orthotopic xenograft (PDOX) models and eight matched cell lines from diverse groups of pHGG. These models recapitulate histopathology, DNA methylation signatures, mutations and gene expression patterns of the patient tumors from which they were derived, and include rare subgroups not well-represented by existing models. We deploy 16 new and existing cell lines for high-throughput screening (HTS). In vitro HTS results predict variable in vivo response to PI3K/mTOR and MEK pathway inhibitors. These unique new models and an online interactive data portal for exploration of associated detailed molecular characterization and HTS chemical sensitivity data provide a rich resource for pediatric brain tumor research.
Collapse
Affiliation(s)
- Chen He
- Department of Developmental Neurobiology, Memphis, TN, USA
| | - Ke Xu
- Center for Applied Bioinformatics, Memphis, TN, USA
- Department of Computational Biology, Memphis, TN, USA
| | - Xiaoyan Zhu
- Department of Developmental Neurobiology, Memphis, TN, USA
| | - Paige S Dunphy
- Department of Developmental Neurobiology, Memphis, TN, USA
- Department of Oncology, Memphis, TN, USA
| | - Brian Gudenas
- Department of Developmental Neurobiology, Memphis, TN, USA
| | - Wenwei Lin
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA
| | - Nathaniel Twarog
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA
| | - Laura D Hover
- Department of Developmental Neurobiology, Memphis, TN, USA
| | | | | | - Junyuan Zhang
- Department of Developmental Neurobiology, Memphis, TN, USA
| | - Xiaoyu Li
- Department of Pathology, Memphis, TN, USA
| | | | | | | | - Duane G Currier
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA
| | - William Caufield
- Preclinical Pharmacokinetics Shared Resource St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yingzhe Wang
- Preclinical Pharmacokinetics Shared Resource St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jia Xie
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA
| | - Alberto Broniscer
- Division of Hematology-Oncology, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | - Paul Klimo
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Frederick Boop
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jinghui Zhang
- Department of Computational Biology, Memphis, TN, USA
| | | | | | - Michelle Monje
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Burgess B Freeman Iii
- Preclinical Pharmacokinetics Shared Resource St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA
| | - Gang Wu
- Center for Applied Bioinformatics, Memphis, TN, USA
- Department of Computational Biology, Memphis, TN, USA
| | | | - Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, Memphis, TN, USA.
| | | |
Collapse
|
7
|
Singh P, Kumar V, Gupta SK, Kumari G, Verma M. Combating TKI resistance in CML by inhibiting the PI3K/Akt/mTOR pathway in combination with TKIs: a review. Med Oncol 2021; 38:10. [PMID: 33452624 DOI: 10.1007/s12032-021-01462-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/03/2021] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML), a myeloproliferative hematopoietic cancer, is caused by a genetic translocation between chromosomes 9 and 22. This translocation produces a small Philadelphia chromosome, which contains the Bcr-Abl oncogene. The Bcr-Abl oncogene encodes the BCR-ABL protein, upregulates various signaling pathways (JAK-STAT, MAPK/ERK, and PI3K/Akt/mTOR), and out of which the specifically highly active pathway is the PI3K/Akt/mTOR pathway. Among early treatments for CML, tyrosine kinase inhibitors (TKIs) were found to be the most effective, but drug resistance against kinase inhibitors led to the discovery of novel alternative therapies. At this point, the PI3K/Akt/mTOR pathway components became new targets due to stimulation of this pathway in TKIs-resistant CML patients. The current review article deals with reviewing the scientific literature on the PI3K/Akt/mTOR pathway inhibitors listed in the National Cancer Institute (NCI) drug dictionary and proved effective against multiple cancers. And out of those enlisted inhibitors, the US FDA has also approved some PI3K inhibitors (Idelalisib, Copanlisib, and Duvelisib) and mTOR inhibitors (Everolimus, Sirolimus, and Temsirolimus) for cancer therapy. So far, several inhibitors have been tested, and further investigations are still ongoing. Even in Imatinib, Nilotinib, and Ponatinib-resistant CML cells, a dual PI3K/mTOR inhibitor, BEZ235, showed antiproliferative activity. Therefore, by considering the literature data of these reviews and further examining some of the reported inhibitors, which proved effective against the PI3K/Akt/mTOR signaling pathway in multiple cancers, may improve the therapeutic approaches towards TKI-resistant CML cells where the respective signaling pathway gets upregulated.
Collapse
Affiliation(s)
- Priyanka Singh
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Veerandra Kumar
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Sonu Kumar Gupta
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Gudia Kumari
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Bathinda, 151001, India
| | - Malkhey Verma
- Department of Biochemistry, School of Basic & Applied Sciences, Central University of Punjab, Bathinda, 151001, India. .,School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
8
|
Shi Y, Huang C, Yi H, Cao Q, Zhao Y, Chen J, Chen X, Pollock C. RIPK3 blockade attenuates kidney fibrosis in a folic acid model of renal injury. FASEB J 2020; 34:10286-10298. [PMID: 32542792 DOI: 10.1096/fj.201902544rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
Renal fibrosis is common to all forms of progressive kidney disease. However, current therapies to limit renal fibrosis are largely ineffective. Phosphorylation of receptor-interacting serine/threonine-protein kinase (RIPK) 3 has been recently suggested to be a key regulator of the pyrin domain containing 3 (NLRP3) inflammasome, which provides new insights into mechanisms of chronic kidney disease (CKD). However, the specific effect of RIPK3 on renal cortical fibrosis has not been fully understood. To study the function of RIPK3, both genetic ablation and pharmacological inhibition of RIPK3 (dabrafenib) were used in the study. Our studies identify that RIPK3 promotes renal fibrosis via the activation of the NLRP3 inflammasome in a mouse model of folic acid-induced nephropathy. Both interventional strategies decreased the renal fibrotic response, and beneficial effects converged on the NLRP3 inflammasome. This study demonstrates a role for RIPK3 as the mediator of renal fibrosis via the upregulation of inflammasome activation. Dabrafenib, as an inhibitor of RIPK3, may be an effective treatment to limit the progression of the tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Ying Shi
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Chunling Huang
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Hao Yi
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Qinghua Cao
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Yongli Zhao
- Department of Pediatrics, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jason Chen
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Xinming Chen
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Carol Pollock
- Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Xiao Y, Yu Y, Jiang P, Li Y, Wang C, Zhang R. The PI3K/mTOR dual inhibitor GSK458 potently impedes ovarian cancer tumorigenesis and metastasis. Cell Oncol (Dordr) 2020; 43:669-680. [PMID: 32382996 DOI: 10.1007/s13402-020-00514-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The PI3K/AKT/mTOR pathway is one of the most highly activated cellular signaling pathways in advanced ovarian cancer. Although several PI3K/AKT/mTOR inhibitors have been developed to treat various types of cancer, the antitumor efficacy of many of these compounds against ovarian cancer has remained unclear. METHODS Here, we tested and compared a panel of 16 PI3K/AKT/mTOR inhibitors (XL765, Miltefosine, Rapamycin, CCI-779, RAD001, FK506, XL147, GSK2110183, IPI-145, GSK2141795, BYL719, GSK458, CAL-101, XL765 analogue SAR245409, Triciribine, and GDC0941) that have entered clinical trials for antitumor activity against ovarian cancer, as well as the front line drug, paclitaxel. Antitumor efficacy was measured in both ovarian cancer cell lines and patient-derived ovarian primary tumor cell lines in vitro and in vivo. RESULTS We identified the PI3K/mTOR dual inhibitor GSK458 as a potent inhibitor of proliferation in all cell lines tested at half maximal inhibitory concentrations (IC50) of approximately 0.01-1 µM, a range tens to hundreds fold lower than that of the other PI3K/AKT/mTOR inhibitors tested. Additionally, GSK458 showed the highest inhibitory efficacy against ovarian cancer cell migration. GSK458 also inhibited tumor growth and metastasis in nude mice intraperitoneally engrafted with SKOV3 cells or a patient-derived tumor cell xenograft (PDCX). Importantly, the inhibitory efficiency of GSK458 on cell proliferation and migration both in vitro and in vivo was comparable to that of paclitaxel. Mechanistically, the anti-tumor activity of GSK458 was found to be associated with inactivation of AKT and mTOR, and induction of cell cycle arrest at the G0/G1 phase. CONCLUSIONS Based on our results, we conclude that GSK458 may serve as an attractive candidate to treat ovarian cancer.
Collapse
Affiliation(s)
- Yangjiong Xiao
- Department of Obstetrics and Gynecology, Shanghai Fengxian District Central Hospital, Southern Medical University, 201499, Shanghai, China. .,State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 510182, Guangzhou, China. .,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Yang Yu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Pengcheng Jiang
- Department of Gynecology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, 213004, China
| | - Yuhong Li
- Department of Gynecology, The International Peace Maternity & Child Health Hospital, The China Welfare Institute, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Chao Wang
- Department of Gynecology, The International Peace Maternity & Child Health Hospital, The China Welfare Institute, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Rong Zhang
- Department of Obstetrics and Gynecology, Shanghai Fengxian District Central Hospital, Southern Medical University, 201499, Shanghai, China.
| |
Collapse
|
10
|
Quan X, Du H, Xu J, Hou X, Gong X, Wu Y, Zhou Y, Jiang J, Lu L, Yuan S, Yang X, Shi L, Sun L. Novel Quinoline Compound Derivatives of NSC23925 as Potent Reversal Agents Against P-Glycoprotein-Mediated Multidrug Resistance. Front Chem 2020; 7:820. [PMID: 31921759 PMCID: PMC6931887 DOI: 10.3389/fchem.2019.00820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/12/2019] [Indexed: 12/02/2022] Open
Abstract
Multidrug resistance is a serious problem and a common cause of cancer treatment failure, leading to patient death. Although numerous reversal resistance inhibitors have been evaluated in preclinical or clinical trials, efficient and low-toxicity reversal agents have not been identified. In this study, a series of novel quinoline compound derivatives from NSC23925 were designed to inhibit P-glycoprotein (P-gp). Among them, YS-7a showed a stronger inhibitory effect against P-gp than verapamil, as a positive control, when co-incubated with chemotherapy drugs at minimally cytotoxic concentrations. YS-7a suppressed the P-gp transport function without affecting the expression of P-gp but stimulated the ATPase activity of P-gp in a dose-dependent manner. Next, molecular docking was used to predict the six most probable binding sites, namely, SER270, VAL273, VAL274, ILE354, VAL357, and PHE390. Moreover, YS-7a had no effect on cytochrome P450 3A4 activity and showed little toxicity to normal cells. In addition, combined treatment of YS-7a with vincristine showed a better inhibitory effect than the positive control verapamil in vivo without a negative effect on mouse weight. Overall, our results showed that YS-7a could reverse cancer multidrug resistance through the inhibition of P-gp transport function in vitro and in vivo, suggesting that YS-7a may be a novel therapeutic agent.
Collapse
Affiliation(s)
- Xingping Quan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jingjing Xu
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Xiaoying Hou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaofeng Gong
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yao Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yuqi Zhou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiangyu Yang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Lei Shi
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Silva D, Sharma M, Barnett GH. Laser Ablation vs Open Resection for Deep-Seated Tumors: Evidence for Laser Ablation. Neurosurgery 2018; 63 Suppl 1:15-26. [PMID: 27399359 DOI: 10.1227/neu.0000000000001289] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Danilo Silva
- Department of Neurosurgery, Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mayur Sharma
- Department of Neurosurgery, Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| | - Gene H Barnett
- Department of Neurosurgery, Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
12
|
Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer 2018; 18:452-464. [PMID: 29643473 PMCID: PMC6622180 DOI: 10.1038/s41568-018-0005-8] [Citation(s) in RCA: 1254] [Impact Index Per Article: 179.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Most patients who die of cancer have disseminated disease that has become resistant to multiple therapeutic modalities. Ample evidence suggests that the expression of ATP-binding cassette (ABC) transporters, especially the multidrug resistance protein 1 (MDR1, also known as P-glycoprotein or P-gp), which is encoded by ABC subfamily B member 1 (ABCB1), can confer resistance to cytotoxic and targeted chemotherapy. However, the development of MDR1 as a therapeutic target has been unsuccessful. At the time of its discovery, appropriate tools for the characterization and clinical development of MDR1 as a therapeutic target were lacking. Thirty years after the initial cloning and characterization of MDR1 and the implication of two additional ABC transporters, the multidrug resistance-associated protein 1 (MRP1; encoded by ABCC1)), and ABCG2, in multidrug resistance, interest in investigating these transporters as therapeutic targets has waned. However, with the emergence of new data and advanced techniques, we propose to re-evaluate whether these transporters play a clinical role in multidrug resistance. With this Opinion article, we present recent evidence indicating that it is time to revisit the investigation into the role of ABC transporters in efficient drug delivery in various cancer types and at the blood-brain barrier.
Collapse
Affiliation(s)
- Robert W Robey
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kristen M Pluchino
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Antonio T Fojo
- Division of Hematology/Oncology, Department of Medicine, Columbia University/New York Presbyterian Hospital, Manhattan, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Susan E Bates
- Division of Hematology/Oncology, Department of Medicine, Columbia University/New York Presbyterian Hospital, Manhattan, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
13
|
Therapeutic Potential and Utility of Elacridar with Respect to P-glycoprotein Inhibition: An Insight from the Published In Vitro, Preclinical and Clinical Studies. Eur J Drug Metab Pharmacokinet 2018; 42:915-933. [PMID: 28374336 DOI: 10.1007/s13318-017-0411-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The occurrence of efflux mechanisms via Permeability-glycoprotein (P-gp) recognized as an important physiological process impedes drug entry or transport across membranes into tissues. In some instances, either low oral bioavailability or lack of brain penetration has been attributed to P-gp mediated efflux activity. Therefore, the objective of development of P-gp inhibitors was to facilitate the attainment of higher drug exposures in tissues. Many third-generation P-gp inhibitors such as elacridar, tariquidar, zosuquidar, etc. have entered clinical development to fulfil the promise. The body of evidence from in vitro and in vivo preclinical and clinical data reviewed in this paper provides the basis for an effective blockade of P-gp efflux mechanism by elacridar. However, clinical translation of the promise has been elusive not just for elacridar but also for other P-gp inhibitors in this class. The review provides introspection and perspectives on the lack of clinical translation of this class of drugs and a broad framework of strategies and considerations in the potential application of elacridar and other P-gp inhibitors in oncology therapeutics.
Collapse
|
14
|
Laramy JK, Kim M, Parrish KE, Sarkaria JN, Elmquist WF. Pharmacokinetic Assessment of Cooperative Efflux of the Multitargeted Kinase Inhibitor Ponatinib Across the Blood-Brain Barrier. J Pharmacol Exp Ther 2018; 365:249-261. [PMID: 29440450 PMCID: PMC5878676 DOI: 10.1124/jpet.117.246116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/08/2018] [Indexed: 12/31/2022] Open
Abstract
A compartmental blood-brain barrier (BBB) model describing drug transport across the BBB was implemented to evaluate the influence of efflux transporters on the rate and extent of the multikinase inhibitor ponatinib penetration across the BBB. In vivo pharmacokinetic studies in wild-type and transporter knockout mice showed that two major BBB efflux transporters, P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp), cooperate to modulate the brain exposure of ponatinib. The total and unbound (free) brain-to-plasma ratios were approximately 15-fold higher in the triple knockout mice lacking both P-gp and Bcrp [Mdr1a/b(-/-)Bcrp1(-/-)] compared with the wild-type mice. The triple knockout mice had a greater than an additive increase in the brain exposure of ponatinib when compared with single knockout mice [Bcrp1(-/-) or Mdr1a/b(-/-)], suggesting functional compensation of transporter-mediated drug efflux. Based on the BBB model characterizing the observed brain and plasma concentration-time profiles, the brain exit rate constant and clearance out of the brain were approximately 15-fold higher in the wild-type compared with Mdr1a/b(-/-)Bcrp1(-/-) mice, resulting in a significant increase in the mean transit time (the average time spent by ponatinib in the brain in a single passage) in the absence of efflux transporters (P-gp and Bcrp). This study characterized transporter-mediated drug efflux from the brain, a process that reduces the duration and extent of ponatinib exposure in the brain and has critical implications for the use of targeted drug delivery for brain tumors.
Collapse
Affiliation(s)
- Janice K Laramy
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J.K.L., M.K., K.E.P., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Minjee Kim
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J.K.L., M.K., K.E.P., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Karen E Parrish
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J.K.L., M.K., K.E.P., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - Jann N Sarkaria
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J.K.L., M.K., K.E.P., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J.K.L., M.K., K.E.P., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.)
| |
Collapse
|
15
|
Westphal D, Glitza Oliva IC, Niessner H. Molecular insights into melanoma brain metastases. Cancer 2017; 123:2163-2175. [PMID: 28543697 DOI: 10.1002/cncr.30594] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/19/2016] [Accepted: 12/28/2016] [Indexed: 01/26/2023]
Abstract
Substantial proportions of patients with metastatic melanoma develop brain metastases during the course of their disease, often resulting in significant morbidity and death. Despite recent advances with BRAF/MEK and immune-checkpoint inhibitors in the treatment of patients who have melanoma with extracerebral metastases, patients who have melanoma brain metastases still have poor overall survival, highlighting the need for further therapy options. A deeper understanding of the molecular pathways involved in the development of melanoma brain metastases is required to develop more brain-specific therapies. Here, the authors summarize the currently known preclinical data and describe steps involved in the development of melanoma brain metastases. Only by knowing the molecular background is it possible to design new therapeutic agents that can be used to improve the outcome of patients with melanoma brain metastases. Cancer 2017;123:2163-75. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Dana Westphal
- Department of Dermatology, Carl Gustav Carus Medical Center, Technical University of Dresden, Dresden, Germany.,Center for Regenerative Therapies, Technical University of Dresden, Dresden, Germany
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heike Niessner
- Department of Dermatology, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
16
|
Gampa G, Vaidhyanathan S, Sarkaria JN, Elmquist WF. Drug delivery to melanoma brain metastases: Can current challenges lead to new opportunities? Pharmacol Res 2017. [PMID: 28634084 DOI: 10.1016/j.phrs.2017.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Melanoma has a high propensity to metastasize to the brain, and patients with melanoma brain metastases (MBM) have an extremely poor prognosis. The recent approval of several molecularly-targeted agents (e.g., BRAF, MEK inhibitors) and biologics (anti-CTLA-4, anti-PD-1 and anti-PD-L1 antibodies) has brought new hope to patients suffering from this formerly untreatable and lethal disease. Importantly, there have been recent reports of success in some clinical studies examining the efficacy of both targeted agents and immunotherapies that show similar response rates in both brain metastases and extracranial disease. While these studies are encouraging, there remains significant room for improvement in the treatment of MBM, given the lack of durable response and the development of resistance to current therapies. Critical questions remain regarding mechanisms that lead to this lack of durable response and development of resistance, and how those mechanisms may differ in systemic sites versus brain metastases. One issue that may not be fully appreciated is that the delivery of several small molecule molecularly-targeted therapies to the brain is often restricted due to active efflux at the blood-brain barrier (BBB) interface. Inadequate local drug concentrations may be partially responsible for the development of unique patterns of resistance at metastatic sites in the brain. It is clear that there can be local, heterogeneous BBB breakdown in MBM, as exemplified by contrast-enhancement on T1-weighted MR imaging. However, it is possible that the successful treatment of MBM with small molecule targeted therapies will depend, in part, on the ability of these therapies to penetrate an intact BBB and reach the protected micro-metastases (so called "sub-clinical" disease) that escape early detection by contrast-enhanced MRI, as well as regions of tumor within MRI-detectable metastases that may have a less compromised BBB. The emergence of resistance in MBM may be related to several diverse, yet interrelated, factors including the distinct microenvironment of the brain and inadequate brain penetration of targeted therapies to specific regions of tumor. The tumor microenvironment has been ascribed to play a key role in steering the course of disease progression, by dictating changes in expression of tumor drivers and resistance-related signaling mechanisms. Therefore, a key issue to consider is how changes in drug delivery, and hence local drug concentrations within a metastatic microenvironment, will influence the development of resistance. Herein we discuss our perspective on several critical questions that focus on many aspects relevant to the treatment of melanoma brain metastases; the answers to which may lead to important advances in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Gautham Gampa
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Shruthi Vaidhyanathan
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | | | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
17
|
Gampa G, Vaidhyanathan S, Resman BW, Parrish KE, Markovic SN, Sarkaria JN, Elmquist WF. Challenges in the delivery of therapies to melanoma brain metastases. ACTA ACUST UNITED AC 2016; 2:309-325. [PMID: 28546917 DOI: 10.1007/s40495-016-0072-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Brain metastases are a major cause of morbidity and mortality in patients with advanced melanoma. Recent approval of several molecularly-targeted agents and biologics has brought hope to patients with this previously untreatable disease. However, patients with symptomatic melanoma brain metastases have often been excluded from pivotal clinical trials. This may be in part attributed to the fact that several of the approved small molecule molecularly-targeted agents are substrates for active efflux at the blood-brain barrier, limiting their effective delivery to brain metastases. We believe that successful treatment of melanoma brain metastases will depend on the ability of these agents to traverse the blood-brain barrier and reach micrometastases that are often not clinically detectable. Moreover, overcoming the emergence of a unique pattern of resistance, possibly through adequate delivery of combination targeted therapies in brain metastases will be important in achieving a durable response. These concepts, and the current challenges in the delivery of new treatments to melanoma brain metastases, are discussed in this review.
Collapse
Affiliation(s)
- Gautham Gampa
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shruthi Vaidhyanathan
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brynna-Wilken Resman
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Karen E Parrish
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|