1
|
Ha Y, Rajani K, Riviere-Cazaux C, Rahman M, Olson IE, Gharibi Loron A, Schroeder MA, Rodriguez M, Warrington AE, Burns TC. An Injury-like Signature of the Extracellular Glioma Metabolome. Cancers (Basel) 2024; 16:2705. [PMID: 39123433 PMCID: PMC11311774 DOI: 10.3390/cancers16152705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Aberrant metabolism is a hallmark of malignancies including gliomas. Intracranial microdialysis enables the longitudinal collection of extracellular metabolites within CNS tissues including gliomas and can be leveraged to evaluate changes in the CNS microenvironment over a period of days. However, delayed metabolic impacts of CNS injury from catheter placement could represent an important covariate for interpreting the pharmacodynamic impacts of candidate therapies. Intracranial microdialysis was performed in patient-derived glioma xenografts of glioma before and 72 h after systemic treatment with either temozolomide (TMZ) or a vehicle. Microdialysate from GBM164, an IDH-mutant glioma patient-derived xenograft, revealed a distinct metabolic signature relative to the brain that recapitulated the metabolic features observed in human glioma microdialysate. Unexpectedly, catheter insertion into the brains of non-tumor-bearing animals triggered metabolic changes that were significantly enriched for the extracellular metabolome of glioma itself. TMZ administration attenuated this resemblance. The human glioma microdialysate was significantly enriched for both the PDX versus brain signature in mice and the induced metabolome of catheter placement within the murine control brain. These data illustrate the feasibility of microdialysis to identify and monitor the extracellular metabolome of diseased versus relatively normal brains while highlighting the similarity between the extracellular metabolome of human gliomas and that of CNS injury.
Collapse
Affiliation(s)
- Yooree Ha
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Karishma Rajani
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Cecile Riviere-Cazaux
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Masum Rahman
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Ian E. Olson
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60208, USA
| | - Ali Gharibi Loron
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Mark A. Schroeder
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Arthur E. Warrington
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| | - Terry C. Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55905, USA; (Y.H.); (K.R.); (C.R.-C.); (M.R.); (I.E.O.); (A.G.L.); (M.A.S.); (A.E.W.)
| |
Collapse
|
2
|
Lim S, Kwak M, Kang J, Cesaire M, Tang K, Robey RW, Frye WJE, Karim B, Butcher D, Lizak MJ, Dalmage M, Foster B, Nuechterlein N, Eberhart C, Cimino PJ, Gottesman MM, Jackson S. Ibrutinib disrupts blood-tumor barrier integrity and prolongs survival in rodent glioma model. Acta Neuropathol Commun 2024; 12:56. [PMID: 38589905 PMCID: PMC11003129 DOI: 10.1186/s40478-024-01763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/24/2024] [Indexed: 04/10/2024] Open
Abstract
In malignant glioma, cytotoxic drugs are often inhibited from accessing the tumor site due to the blood-tumor barrier (BTB). Ibrutinib, FDA-approved lymphoma agent, inhibits Bruton tyrosine kinase (BTK) and has previously been shown to independently impair aortic endothelial adhesion and increase rodent glioma model survival in combination with cytotoxic therapy. Yet additional research is required to understand ibrutinib's effect on BTB function. In this study, we detail baseline BTK expression in glioma cells and its surrounding vasculature, then measure endothelial junctional expression/function changes with varied ibrutinib doses in vitro. Rat glioma cells and rodent glioma models were treated with ibrutinib alone (1-10 µM and 25 mg/kg) and in combination with doxil (10-100 µM and 3 mg/kg) to assess additive effects on viability, drug concentrations, tumor volume, endothelial junctional expression and survival. We found that ibrutinib, in a dose-dependent manner, decreased brain endothelial cell-cell adhesion over 24 h, without affecting endothelial cell viability (p < 0.005). Expression of tight junction gene and protein expression was decreased maximally 4 h after administration, along with inhibition of efflux transporter, ABCB1, activity. We demonstrated an additive effect of ibrutinib with doxil on rat glioma cells, as seen by a significant reduction in cell viability (p < 0.001) and increased CNS doxil concentration in the brain (56 ng/mL doxil alone vs. 74.6 ng/mL combination, p < 0.05). Finally, Ibrutinib, combined with doxil, prolonged median survival in rodent glioma models (27 vs. 16 days, p < 0.0001) with brain imaging showing a - 53% versus - 75% volume change with doxil alone versus combination therapy (p < 0.05). These findings indicate ibrutinib's ability to increase brain endothelial permeability via junctional disruption and efflux inhibition, to increase BTB drug entry and prolong rodent glioma model survival. Our results motivate the need to identify other BTB modifiers, all with the intent of improving survival and reducing systemic toxicities.
Collapse
Affiliation(s)
- Sanghee Lim
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Minhye Kwak
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jeonghan Kang
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Melissa Cesaire
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Kayen Tang
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - William J E Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory, Leidos Biomedical Research, Frederick, MD, 21702, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory, Leidos Biomedical Research, Frederick, MD, 21702, USA
| | - Martin J Lizak
- NIH MRI Research Facility and Mouse Imaging Facility, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Bethesda, MD, 20814, USA
| | - Mahalia Dalmage
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Brandon Foster
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Nicholas Nuechterlein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles Eberhart
- Neuropathology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Bethesda, MD, 20892, USA
| | - Patrick J Cimino
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, 20892, USA
| | - Sadhana Jackson
- Develomental Therapeutics and Pharmacology Unit, Surgical Neurology Branch, National Institute of Neurologic Disorders and Stroke (NINDS), NIH, Building 10, Room 7D45, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
3
|
Virtanen PS, Ortiz KJ, Patel A, Blocher WA, Richardson AM. Blood-Brain Barrier Disruption for the Treatment of Primary Brain Tumors: Advances in the Past Half-Decade. Curr Oncol Rep 2024; 26:236-249. [PMID: 38329660 DOI: 10.1007/s11912-024-01497-7] [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] [Accepted: 12/28/2023] [Indexed: 02/09/2024]
Abstract
PURPOSE OF REVIEW To review relevant advances in the past half-decade in the treatment of primary brain tumors via modification of blood-brain barrier (BBB) permeability. RECENT FINDINGS BBB disruption is becoming increasingly common in the treatment of primary brain tumors. Use of mannitol in BBB disruption for targeted delivery of chemotherapeutics via superselective intra-arterial cerebral infusion (SIACI) is the most utilized strategy to modify the BBB. Mannitol is used in conjunction with chemotherapeutics, oligonucleotides, and other active agents. Convection-enhanced delivery has become an attractive option for therapeutic delivery while bypassing the BBB. Other technologic innovations include laser interstitial thermal therapy (LITT) and focused ultrasound (FUS) which have emerged as prime modalities to directly target tumors and cause significant local BBB disruption. In the past 5 years, interest has significantly increased in studying modalities to disrupt the BBB in primary brain tumors to enhance treatment responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Piiamaria S Virtanen
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kyle J Ortiz
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ajay Patel
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Angela M Richardson
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
4
|
Ducray F, Ramirez C, Robert M, Fontanilles M, Bronnimann C, Chinot O, Estrade F, Durando X, Cartalat S, Bastid J, Bienayme H, Lemarchand C. A Multicenter Randomized Bioequivalence Study of a Novel Ready-to-Use Temozolomide Oral Suspension vs. Temozolomide Capsules. Pharmaceutics 2023; 15:2664. [PMID: 38140005 PMCID: PMC10747054 DOI: 10.3390/pharmaceutics15122664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Temozolomide (TMZ) oral suspension (Ped-TMZ, KIZFIZO®) is being developed for the treatment of relapsed or refractory neuroblastoma, a rare cancer affecting infants and young children. The study assessed the safety and the bioequivalence of this novel pediatric formulation with existing TMZ oral capsules. METHODS In vitro dissolution profiles and the bioequivalence were evaluated following the European Medicines Agency "Guidelines on the investigation of Bioequivalence". The phase I, multicenter, randomized, open-label, crossover, single-dose bioequivalence study enrolled 36 adult patients with glioblastoma multiforme or lower-grade glioma. Each patient received 200 mg/m2 Ped-TMZ suspension and TMZ capsules (Temodal®) on 2 consecutive days, with the order being randomly assigned. Fourteen blood samples were collected up to 10 h post-dosing. Bioequivalence was assessed by comparing the 90% confidence interval for the ratio of the geometric means of maximum TMZ plasma concentration (Cmax) and the area under the curve (AUCt). Other endpoints included further pharmacokinetic parameters and safety. RESULTS Both formulations exhibited a fast in vitro dissolution profile with more than 85% of TMZ dissolved within 15 min. For the bioequivalence study, thirty patients completed the trial as per the protocol. The ratio of Ped-TMZ/TMZ capsule geometric means (90% CI) for AUCt and Cmax were 97.18% (95.05-99.35%) and 107.62% (98.07-118.09%), respectively, i.e., within the 80-125% bioequivalence limits. No buccal toxicity was associated with Ped-TMZ liquid formulation. CONCLUSIONS This study showed that Ped-TMZ oral suspension and TMZ oral capsule treatment are immediate release and bioequivalent medicines. There were also no unexpected safety signals or local toxicity (funded by ORPHELIA Pharma; ClinicalTrials.gov number, NCT04467346).
Collapse
Affiliation(s)
- François Ducray
- Service de Neuro-Oncologie, Hôpital Neurologique, Hospices Civils de Lyon, Centre de Recherche en Cancérologie UMR INSERM 1052 CNRS 5286, Université Claude Bernard Lyon 1, 69008 Lyon, France;
| | - Carole Ramirez
- Services de Neurologie et D’oncologie Médicale, CHU et ICHUSE de Saint-Etienne, 42055 Saint-Etienne, France;
| | - Marie Robert
- Institut de Cancérologie de l’Ouest, Medical Oncology, 44800 Saint Herblain, France;
| | - Maxime Fontanilles
- INSERM U1245 Unit, Cancer Centre Henri Becquerel, Université Rouen Normandie, 76038 Rouen, France;
- Le Havre Hospital Group, 76083 Le Havre, France
| | - Charlotte Bronnimann
- CHU de Bordeaux, Service D’oncologie Médicale, Hôpital Saint André, 33075 Bordeaux, France;
| | - Olivier Chinot
- Aix-Marseille Université, Neuro-Oncology Department, APHM, CNRS, Institut de Neurophysiopathologie, CHU Timone, Service de Neuro-Oncologie, 13385 Marseille, France;
| | | | - Xavier Durando
- INSERM U1240 IMoST, University of Clermont Auvergne, 63001 Clermont-Ferrand, France;
- UMR 501, Clinical Investigation Centre, 63011 Clermont-Ferrand, France
- Clinical Research and Innovation Department, Centre Jean Perrin, 63011 Clermont-Ferrand, France
- Oncology Department, Centre Jean Perrin, 63011 Clermont-Ferrand, France
| | - Stéphanie Cartalat
- Service de Neuro-Oncologie, Hôpital Neurologique, Hospices Civils de Lyon, Centre de Recherche en Cancérologie UMR INSERM 1052 CNRS 5286, Université Claude Bernard Lyon 1, 69008 Lyon, France;
| | - Jeremy Bastid
- ORPHELIA Pharma, 75005 Paris, France; (J.B.); (H.B.)
| | | | | |
Collapse
|
5
|
Fernandez M, Nigro M, Travagli A, Pasquini S, Vincenzi F, Varani K, Borea PA, Merighi S, Gessi S. Strategies for Drug Delivery into the Brain: A Review on Adenosine Receptors Modulation for Central Nervous System Diseases Therapy. Pharmaceutics 2023; 15:2441. [PMID: 37896201 PMCID: PMC10610137 DOI: 10.3390/pharmaceutics15102441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a biological barrier that protects the central nervous system (CNS) by ensuring an appropriate microenvironment. Brain microvascular endothelial cells (ECs) control the passage of molecules from blood to brain tissue and regulate their concentration-versus-time profiles to guarantee proper neuronal activity, angiogenesis and neurogenesis, as well as to prevent the entry of immune cells into the brain. However, the BBB also restricts the penetration of drugs, thus presenting a challenge in the development of therapeutics for CNS diseases. On the other hand, adenosine, an endogenous purine-based nucleoside that is expressed in most body tissues, regulates different body functions by acting through its G-protein-coupled receptors (A1, A2A, A2B and A3). Adenosine receptors (ARs) are thus considered potential drug targets for treating different metabolic, inflammatory and neurological diseases. In the CNS, A1 and A2A are expressed by astrocytes, oligodendrocytes, neurons, immune cells and ECs. Moreover, adenosine, by acting locally through its receptors A1 and/or A2A, may modulate BBB permeability, and this effect is potentiated when both receptors are simultaneously activated. This review showcases in vivo and in vitro evidence supporting AR signaling as a candidate for modifying endothelial barrier permeability in the treatment of CNS disorders.
Collapse
Affiliation(s)
- Mercedes Fernandez
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| | - Manuela Nigro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| | - Alessia Travagli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| | - Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy;
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| | | | - Stefania Merighi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| | - Stefania Gessi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (M.F.); (M.N.); (A.T.); (F.V.); (K.V.)
| |
Collapse
|
6
|
Riviere-Cazaux C, Neth BJ, Hoplin MD, Wessel B, Miska J, Kizilbash SH, Burns TC. Glioma Metabolic Feedback In Situ: A First-In-Human Pharmacodynamic Trial of Difluoromethylornithine + AMXT-1501 Through High-Molecular Weight Microdialysis. Neurosurgery 2023; 93:932-938. [PMID: 37246885 PMCID: PMC10637404 DOI: 10.1227/neu.0000000000002511] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND AND OBJECTIVES No new drug has improved survival for glioblastoma since temozolomide in 2005, due in part to the relative inaccessibility of each patient's individualized tumor biology and its response to therapy. We have identified a conserved extracellular metabolic signature of enhancing high-grade gliomas enriched for guanidinoacetate (GAA). GAA is coproduced with ornithine, the precursor to protumorigenic polyamines through ornithine decarboxylase (ODC). AMXT-1501 is a polyamine transporter inhibitor that can overcome tumoral resistance to the ODC inhibitor, difluoromethylornithine (DFMO). We will use DFMO with or without AMXT-1501 to identify candidate pharmacodynamic biomarkers of polyamine depletion in patients with high-grade gliomas in situ . We aim to determine (1) how blocking polyamine production affects intratumoral extracellular guanidinoacetate abundance and (2) the impact of polyamine depletion on the global extracellular metabolome within live human gliomas in situ. METHODS DFMO, with or without AMXT-1501, will be administered postoperatively in 15 patients after clinically indicated subtotal resection for high-grade glioma. High-molecular weight microdialysis catheters implanted into residual tumor and adjacent brain will be used for postoperative monitoring of extracellular GAA and polyamines throughout therapeutic intervention from postoperative day (POD) 1 to POD5. Catheters will be removed on POD5 before discharge. EXPECTED OUTCOMES We anticipate that GAA will be elevated in tumor relative to adjacent brain although it will decrease within 24 hours of ODC inhibition with DFMO. If AMXT-1501 effectively increases the cytotoxic impact of ODC inhibition, we expect an increase in biomarkers of cytotoxicity including glutamate with DFMO + AMXT-1501 treatment when compared with DFMO alone. DISCUSSION Limited mechanistic feedback from individual patients' gliomas hampers clinical translation of novel therapies. This pilot Phase 0 study will provide in situ feedback during DFMO + AMXT-1501 treatment to determine how high-grade gliomas respond to polyamine depletion.
Collapse
Affiliation(s)
| | - Bryan J. Neth
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew D. Hoplin
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Bambi Wessel
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, Chicago, Illinois, USA
| | | | - Terry C. Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Nduom EK, Glod J, Brown DA, Fagan M, Dalmage M, Heiss J, Steinberg SM, Peer C, Figg WD, Jackson S. Clinical protocol: Feasibility of evaluating abemaciclib neuropharmacokinetics of diffuse midline glioma using intratumoral microdialysis. PLoS One 2023; 18:e0291068. [PMID: 37682953 PMCID: PMC10490936 DOI: 10.1371/journal.pone.0291068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023] Open
Abstract
Diffuse midline gliomas (DMG) are the most aggressive brain tumors of childhood and young adults, with documented 2-year survival rates <10%. Treatment failure is due in part to the function of the BBB. Intratumoral microdialysis sampling is an effective tool to determine brain entry of varied agents and could help to provide a better understanding of the relationship of drug permeability to DMG treatment responsivity. This is a non-randomized, single-center, phase 1 clinical trial. Up to seven young adult (18-39 years) patients with recurrent high-grade or diffuse midline glioma will be enrolled with the goal of 5 patients completing the trial over an anticipated 24 months. All patients will take abemaciclib pre-operatively for 4.5 days at twice daily dosing. Patients will undergo resection or biopsy, placement of a microdialysis catheter, and 48 hours of dialysate sampling coupled with timed plasma collections. If intratumoral tumor or brain dialysate sampling concentrations are >10nmol/L, or tumor tissue studies demonstrate CDK inhibition, then restart of abemaciclib therapy along with temozolomide will be administered for maintenance therapy and discontinued with evidence of radiologic or clinical disease progression. The poor survival associated with diffuse midline gliomas underscore the need for improved means to evaluate efficacy of drug delivery to tumor and peritumoral tissue. The findings of this novel study, will provide real-time measurements of BBB function which have the potential to influence future prognostic and diagnostic decisions in such a lethal disease with limited treatment options. Trial registration: Clinicaltrials.gov, NCT05413304. Registered June 10, 2022, Abemaciclib Neuropharmacokinetics of Diffuse Midline Glioma Using Intratumoral Microdialysis.
Collapse
Affiliation(s)
- Edjah K. Nduom
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - John Glod
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Desmond A. Brown
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Margaret Fagan
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Mahalia Dalmage
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - John Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Cody Peer
- Clinical Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - William D. Figg
- Clinical Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Sadhana Jackson
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States of America
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
8
|
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: 7] [Impact Index Per Article: 7.0] [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
|
9
|
Nhàn NTT, Yamada T, Yamada KH. Peptide-Based Agents for Cancer Treatment: Current Applications and Future Directions. Int J Mol Sci 2023; 24:12931. [PMID: 37629112 PMCID: PMC10454368 DOI: 10.3390/ijms241612931] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Peptide-based strategies have received an enormous amount of attention because of their specificity and applicability. Their specificity and tumor-targeting ability are applied to diagnosis and treatment for cancer patients. In this review, we will summarize recent advancements and future perspectives on peptide-based strategies for cancer treatment. The literature search was conducted to identify relevant articles for peptide-based strategies for cancer treatment. It was performed using PubMed for articles in English until June 2023. Information on clinical trials was also obtained from ClinicalTrial.gov. Given that peptide-based strategies have several advantages such as targeted delivery to the diseased area, personalized designs, relatively small sizes, and simple production process, bioactive peptides having anti-cancer activities (anti-cancer peptides or ACPs) have been tested in pre-clinical settings and clinical trials. The capability of peptides for tumor targeting is essentially useful for peptide-drug conjugates (PDCs), diagnosis, and image-guided surgery. Immunomodulation with peptide vaccines has been extensively tested in clinical trials. Despite such advantages, FDA-approved peptide agents for solid cancer are still limited. This review will provide a detailed overview of current approaches, design strategies, routes of administration, and new technological advancements. We will highlight the success and limitations of peptide-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL 60607, USA
| | - Kaori H. Yamada
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology & Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
10
|
Ahmed MH, Canney M, Carpentier A, Idbaih A. Overcoming the blood brain barrier in glioblastoma: Status and future perspective. Rev Neurol (Paris) 2023; 179:430-436. [PMID: 37062676 DOI: 10.1016/j.neurol.2023.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 03/19/2023] [Indexed: 04/18/2023]
Abstract
Glioblastoma is the most common primary brain malignancy in adults. Treatment of glioblastoma patients is based on neurosurgery, radiation therapy and chemotherapy. Despite this multimodal therapeutic regimen, the prognosis of glioblastoma patients is poor. Indeed, glioblastoma is very resistant to treatments due to multiple molecular and cellular mechanisms including the existence of the blood-brain barrier (BBB). The BBB consists of multiple layers surrounding brain vessels and limits drug penetration within the brain. Therefore, overcoming the BBB is a strategy to increase bioavailability and efficacy of therapeutic agents against glioblastoma cells. The development of two approaches is ongoing: i) enhancing the delivery of drugs to the brain and ii) improving the penetration of drugs into the brain. One way to enhance drug delivery to the brain is through high-dose intravenous chemotherapy, with or without bone marrow transplantation, or via intra-arterial chemotherapy, with or without disrupting the BBB through osmotic means. Conversely, improving drug penetration within the brain can be achieved through modifying either the drug itself or the BBB. Promising results in terms of safety and signals of efficacy were obtained with these approaches in early phase clinical trials. More advanced comparative clinical trials are needed to investigate the clinical benefit for glioblastoma patients.
Collapse
Affiliation(s)
- M H Ahmed
- School of Cancer & Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom
| | | | - A Carpentier
- Sorbonne Université, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurochirurgie, 75013, Paris, France
| | - A Idbaih
- Service de Neurologie 2-Mazarin, Charles Foix, DMU Neurosciences, Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75013 Paris, France.
| |
Collapse
|
11
|
Hashimoto Y, Greene C, Munnich A, Campbell M. The CLDN5 gene at the blood-brain barrier in health and disease. Fluids Barriers CNS 2023; 20:22. [PMID: 36978081 PMCID: PMC10044825 DOI: 10.1186/s12987-023-00424-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
The CLDN5 gene encodes claudin-5 (CLDN-5) that is expressed in endothelial cells and forms tight junctions which limit the passive diffusions of ions and solutes. The blood-brain barrier (BBB), composed of brain microvascular endothelial cells and associated pericytes and end-feet of astrocytes, is a physical and biological barrier to maintain the brain microenvironment. The expression of CLDN-5 is tightly regulated in the BBB by other junctional proteins in endothelial cells and by supports from pericytes and astrocytes. The most recent literature clearly shows a compromised BBB with a decline in CLDN-5 expression increasing the risks of developing neuropsychiatric disorders, epilepsy, brain calcification and dementia. The purpose of this review is to summarize the known diseases associated with CLDN-5 expression and function. In the first part of this review, we highlight the recent understanding of how other junctional proteins as well as pericytes and astrocytes maintain CLDN-5 expression in brain endothelial cells. We detail some drugs that can enhance these supports and are being developed or currently in use to treat diseases associated with CLDN-5 decline. We then summarise mutagenesis-based studies which have facilitated a better understanding of the physiological role of the CLDN-5 protein at the BBB and have demonstrated the functional consequences of a recently identified pathogenic CLDN-5 missense mutation from patients with alternating hemiplegia of childhood. This mutation is the first gain-of-function mutation identified in the CLDN gene family with all others representing loss-of-function mutations resulting in mis-localization of CLDN protein and/or attenuated barrier function. Finally, we summarize recent reports about the dosage-dependent effect of CLDN-5 expression on the development of neurological diseases in mice and discuss what cellular supports for CLDN-5 regulation are compromised in the BBB in human diseases.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| | - Chris Greene
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland
| | - Arnold Munnich
- Institut Imagine, INSERM UMR1163, Université Paris Cité, Paris, F-75015, France
- Departments of Pediatric Neurology and Medical Genetics, Hospital Necker Enfants Malades, Université Paris Cité, Paris, F-75015, France
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| |
Collapse
|
12
|
Jo S, Sun IC, Ahn CH, Lee S, Kim K. Recent Trend of Ultrasound-Mediated Nanoparticle Delivery for Brain Imaging and Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:120-137. [PMID: 35184560 DOI: 10.1021/acsami.1c22803] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In view of the fact that the blood-brain barrier (BBB) prevents the transport of imaging probes and therapeutic agents to the brain and thus hinders the diagnosis and treatment of brain-related disorders, methods of circumventing this problem (e.g., ultrasound-mediated nanoparticle delivery) have drawn much attention. Among the related techniques, focused ultrasound (FUS) is a favorite means of enhancing drug delivery via transient BBB opening. Photoacoustic brain imaging relies on the conversion of light into heat and the detection of ultrasound signals from contrast agents, offering the benefits of high resolution and large penetration depth. The extensive versatility and adjustable physicochemical properties of nanoparticles make them promising therapeutic agents and imaging probes, allowing for successful brain imaging and treatment through the combined action of ultrasound and nanoparticulate agents. FUS-induced BBB opening enables nanoparticle-based drug delivery systems to efficiently access the brain. Moreover, photoacoustic brain imaging using nanoparticle-based contrast agents effectively visualizes brain morphologies or diseases. Herein, we review the progress in the simultaneous use of nanoparticles and ultrasound in brain research, revealing the potential of ultrasound-mediated nanoparticle delivery for the effective diagnosis and treatment of brain disorders.
Collapse
Affiliation(s)
- SeongHoon Jo
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Republic of Korea
- Research Institute of Advanced Materials (RIAM), Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul08826, Republic of Korea
| | - In-Cheol Sun
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Cheol-Hee Ahn
- Research Institute of Advanced Materials (RIAM), Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul08826, Republic of Korea
| | - Sangmin Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul02447, Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
13
|
Nanomedicine approaches for medulloblastoma therapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00597-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
14
|
Wang S, Feng Y, Chen L, Yu J, Van Ongeval C, Bormans G, Li Y, Ni Y. Towards updated understanding of brain metastasis. Am J Cancer Res 2022; 12:4290-4311. [PMID: 36225632 PMCID: PMC9548021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/06/2022] [Indexed: 06/16/2023] Open
Abstract
Brain metastasis (BM) is a common complication in cancer patients with advanced disease and attributes to treatment failure and final mortality. Currently there are several therapeutic options available; however these are only suitable for limited subpopulation: surgical resection or radiosurgery for cases with a limited number of lesions, targeted therapies for approximately 18% of patients, and immune checkpoint inhibitors with a response rate of 20-30%. Thus, there is a pressing need for development of novel diagnostic and therapeutic options. This overview article aims to provide research advances in disease model, targeted therapy, blood brain barrier (BBB) opening strategies, imaging and its incorporation with artificial intelligence, external radiotherapy, and internal targeted radionuclide theragnostics. Finally, a distinct type of BM, leptomeningeal metastasis is also covered.
Collapse
Affiliation(s)
- Shuncong Wang
- KU Leuven, Biomedical Group, Campus GasthuisbergLeuven 3000, Belgium
| | - Yuanbo Feng
- KU Leuven, Biomedical Group, Campus GasthuisbergLeuven 3000, Belgium
| | - Lei Chen
- KU Leuven, Biomedical Group, Campus GasthuisbergLeuven 3000, Belgium
| | - Jie Yu
- KU Leuven, Biomedical Group, Campus GasthuisbergLeuven 3000, Belgium
| | - Chantal Van Ongeval
- Department of Radiology, University Hospitals Leuven, KU LeuvenHerestraat 49, Leuven 3000, Belgium
| | - Guy Bormans
- KU Leuven, Biomedical Group, Campus GasthuisbergLeuven 3000, Belgium
| | - Yue Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health SciencesShanghai 201318, China
| | - Yicheng Ni
- KU Leuven, Biomedical Group, Campus GasthuisbergLeuven 3000, Belgium
| |
Collapse
|
15
|
Smith-Cohn MA, Burley NB, Grossman SA. Transient Opening of the Blood-Brain Barrier by Vasoactive Peptides to Increase CNS Drug Delivery: Reality Versus Wishful Thinking? Curr Neuropharmacol 2022; 20:1383-1399. [PMID: 35100958 PMCID: PMC9881081 DOI: 10.2174/1570159x20999220131163504] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/02/2021] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The blood-brain barrier inhibits the central nervous system penetration of 98% of small molecule drugs and virtually all biologic agents, which has limited progress in treating neurologic disease. Vasoactive peptides have been shown in animal studies to transiently disrupt the blood-brain barrier and regadenoson is currently being studied in humans to determine if it can improve drug delivery to the brain. However, many other vasoactive peptides could potentially be used for this purpose. METHODS We performed a review of the literature evaluating the physiologic effects of vasoactive peptides on the vasculature of the brain and systemic organs. To assess the likelihood that a vasoactive peptide might transiently disrupt the blood-brain barrier, we devised a four-tier classification system to organize the available evidence. RESULTS We identified 32 vasoactive peptides with potential blood-brain barrier permeabilityaltering properties. To date, none of these are shown to open the blood-brain barrier in humans. Twelve vasoactive peptides increased blood-brain barrier permeability in rodents. The remaining 20 had favorable physiologic effects on blood vessels but lacked specific information on permeability changes to the blood-brain barrier. CONCLUSION Vasoactive peptides remain an understudied class of drugs with the potential to increase drug delivery and improve treatment in patients with brain tumors and other neurologic diseases. Dozens of vasoactive peptides have yet to be formally evaluated for this important clinical effect. This narrative review summarizes the available data on vasoactive peptides, highlighting agents that deserve further in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Matthew A. Smith-Cohn
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, USA; ,Address correspondence to these authors at the The Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Swedish Health Services, 500 17th Ave, James Tower, Suite 540, Seattle, WA 98122, USA; Tel: 206-320-2300; Fax: 206-320-8149; E-mail: , Sidney Kimmel Cancer Center, Skip Viragh Building, 201 North Broadway, 9th Floor (Mailbox #3), Baltimore, MD 21287, USA; E-mail:
| | - Nicholas B. Burley
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA;
| | - Stuart A. Grossman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA,Address correspondence to these authors at the The Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Swedish Health Services, 500 17th Ave, James Tower, Suite 540, Seattle, WA 98122, USA; Tel: 206-320-2300; Fax: 206-320-8149; E-mail: , Sidney Kimmel Cancer Center, Skip Viragh Building, 201 North Broadway, 9th Floor (Mailbox #3), Baltimore, MD 21287, USA; E-mail:
| |
Collapse
|
16
|
Neurosurgical Clinical Trials for Glioblastoma: Current and Future Directions. Brain Sci 2022; 12:brainsci12060787. [PMID: 35741672 PMCID: PMC9221299 DOI: 10.3390/brainsci12060787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023] Open
Abstract
The mainstays of glioblastoma treatment, maximal safe resection, radiotherapy preserving neurological function, and temozolomide (TMZ) chemotherapy have not changed for the past 17 years despite significant advances in the understanding of the genetics and molecular biology of glioblastoma. This review highlights the neurosurgical foundation for glioblastoma therapy. Here, we review the neurosurgeon’s role in several new and clinically-approved treatments for glioblastoma. We describe delivery techniques such as blood–brain barrier disruption and convection-enhanced delivery (CED) that may be used to deliver therapeutic agents to tumor tissue in higher concentrations than oral or intravenous delivery. We mention pivotal clinical trials of immunotherapy for glioblastoma and explain their outcomes. Finally, we take a glimpse at ongoing clinical trials and promising translational studies to predict ways that new therapies may improve the prognosis of patients with glioblastoma.
Collapse
|
17
|
Sander T, Ghanawi J, Wilson E, Muhammad S, Macleod M, Kahlert UD. Meta-analysis on reporting practices as a source of heterogeneity in in vitro cancer research. BMJ OPEN SCIENCE 2022; 6:e100272. [PMID: 35721833 PMCID: PMC9171230 DOI: 10.1136/bmjos-2021-100272] [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: 12/14/2021] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Heterogeneity of results of exact same research experiments oppose a significant socioeconomic burden. Insufficient methodological reporting is likely to be one of the contributors to results heterogeneity; however, little knowledge on reporting habits of in vitro cancer research and their effects on results reproducibility is available. Exemplified by a commonly performed in vitro assay, we aim to fill this knowledge gap and to derive recommendations necessary for reproducible, robust and translational preclinical science. Methods Here, we use systematic review to describe reporting practices in in vitro glioblastoma research using the Uppsala-87 Malignant Glioma (U-87 MG) cell line and perform multilevel random-effects meta-analysis followed by meta-regression to explore sources of heterogeneity within that literature, and any associations between reporting characteristics and reported findings. Literature that includes experiments measuring the effect of temozolomide on the viability of U-87 MG cells is searched on three databases (Embase, PubMed and Web of Science). Results In 137 identified articles, the methodological reporting is incomplete, for example, medium glucose level and cell density are reported in only 21.2% and 16.8% of the articles. After adjustments for different drug concentrations and treatment durations, the results heterogeneity across the studies (I2=68.5%) is concerningly large. Differences in culture medium glucose level are a driver of this heterogeneity. However, infrequent reporting of most experimental parameters limits the analysis of reproducibility moderating parameters. Conclusions Our results further support the ongoing efforts of establishing consensus reporting practices to elevate durability of results. By doing so, this work can raise awareness of how stricter reporting may help to improve the frequency of successful translation of preclinical results into human application. The authors received no specific funding for this work. A preregistered protocol is available at the Open Science Framework (https://osf.io/9k3dq).
Collapse
Affiliation(s)
- Timo Sander
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | | | - Emma Wilson
- Centre for Clinical Brain Sciences, The University of Edinburgh Medical School, Edinburgh, UK
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, The University of Edinburgh Medical School, Edinburgh, UK
| | - Ulf Dietrich Kahlert
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular and Transplant Surgery, Otto von Guericke Universität Magdeburg, Magdeburg, Germany
| |
Collapse
|
18
|
Ye X, Schreck KC, Ozer BH, Grossman SA. High-grade glioma therapy: adding flexibility in trial design to improve patient outcomes. Expert Rev Anticancer Ther 2022; 22:275-287. [PMID: 35130447 DOI: 10.1080/14737140.2022.2038138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Outcomes for patients with high grade gliomas have changed little over the past thirty years. This realization prompted renewed efforts to increase flexibility in the design and conduct of clinical brain tumor trials. AREAS COVERED This manuscript reviews the development of clinical trial methods, challenges and considerations of flexible clinical trial designs, approaches to improve identification and testing of active agents for high grade gliomas, and evaluation of their delivery to the central nervous system. EXPERT OPINION Flexibility can be introduced in clinical trials in several ways. Flexible designs tout smaller sample sizes, adaptive modifications, fewer control arms, and inclusion of multiple arms in one study. Unfortunately, modifications in study designs cannot address two challenges that are largely responsible for the lack of progress in treating high grade gliomas: 1) the identification of active pharmaceutical agents and 2) the delivery of these agents to brain tumor tissue in therapeutic concentrations. To improve the outcomes of patients with high grade gliomas efforts must be focused on the pre-clinical screening of drugs for activity, the ability of these agents to achieve therapeutic concentrations in non-enhancing tumors, and a willingness to introduce novel compounds in minimally pre-treated patient populations.
Collapse
Affiliation(s)
- Xiaobu Ye
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Karisa C Schreck
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Byram H Ozer
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| | - Stuart A Grossman
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore MD, USA
| |
Collapse
|
19
|
A scoping review of pediatric microdialysis: A missed opportunity for microdialysis in the pediatric neuro-oncology setting. Neurooncol Adv 2022; 4:vdac171. [DOI: 10.1093/noajnl/vdac171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Abstract
Background
Brain microdialysis is a minimally invasive technique for monitoring analytes, metabolites, drugs, neurotransmitters, and/or cytokines. Studies to date have centered on adults with traumatic brain injury, with a limited number of pediatric studies performed. This scoping review details past use of brain microdialysis in children and identifies potential use for future neuro-oncology trials.
Methods
In December 2020, Cochrane Library: CENTRAL, Embase, PubMed, Scopus, and Web of Science: Core Collection were searched. Two reviewers screened all articles by title and abstract review and then full study texts, using microdialysis in patients less than 18 yo.
Results
Of the 1171 articles screened, 49 were included. The 49 studies included 472 pediatric patients (age range 0–17 years old), in the brain (21), abdominal (16), and musculoskeletal (12) regions. Intracerebral microdialysis was performed in 64 collective patients, with a median age of 11 years old, and predominance in metabolic evaluations.
Conclusion
Historically, pediatric microdialysis was safely performed within the brain in varied neurologic conditions, except neuro-oncology. Adult brain tumor studies using intratumoral/peritumoral microdialysis sampling can inform future pediatric studies to advance diagnosis and treatment options for such aggressive tumors.
Collapse
|
20
|
Comprehensive pharmacogenomics characterization of temozolomide response in gliomas. Eur J Pharmacol 2021; 912:174580. [PMID: 34678239 DOI: 10.1016/j.ejphar.2021.174580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 01/11/2023]
Abstract
Recent developments in pharmacogenomics have created opportunities for predicting temozolomide response in gliomas. Temozolomide is the main first-line alkylating chemotherapeutic drug together with radiotherapy as standard treatments of high-risk gliomas after surgery. However, there are great individual differences in temozolomide response. Besides the heterogeneity of gliomas, pharmacogenomics relevant genetic polymorphisms can not only affect pharmacokinetics of temozolomide but also change anti-tumor effects of temozolomide. This review will summarize pharmacogenomic studies of temozolomide in gliomas which can lay the foundation to personalized chemotherapy.
Collapse
|
21
|
Modulation of the Blood-Brain Barrier for Drug Delivery to Brain. Pharmaceutics 2021; 13:pharmaceutics13122024. [PMID: 34959306 PMCID: PMC8708282 DOI: 10.3390/pharmaceutics13122024] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
The blood-brain barrier (BBB) precisely controls brain microenvironment and neural activity by regulating substance transport into and out of the brain. However, it severely hinders drug entry into the brain, and the efficiency of various systemic therapies against brain diseases. Modulation of the BBB via opening tight junctions, inhibiting active efflux and/or enhancing transcytosis, possesses the potential to increase BBB permeability and improve intracranial drug concentrations and systemic therapeutic efficiency. Various strategies of BBB modulation have been reported and investigated preclinically and/or clinically. This review describes conventional and emerging BBB modulation strategies and related mechanisms, and safety issues according to BBB structures and functions, to try to give more promising directions for designing more reasonable preclinical and clinical studies.
Collapse
|
22
|
Whelan R, Hargaden GC, Knox AJS. Modulating the Blood-Brain Barrier: A Comprehensive Review. Pharmaceutics 2021; 13:1980. [PMID: 34834395 PMCID: PMC8618722 DOI: 10.3390/pharmaceutics13111980] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/23/2022] Open
Abstract
The highly secure blood-brain barrier (BBB) restricts drug access to the brain, limiting the molecular toolkit for treating central nervous system (CNS) diseases to small, lipophilic drugs. Development of a safe and effective BBB modulator would revolutionise the treatment of CNS diseases and future drug development in the area. Naturally, the field has garnered a great deal of attention, leading to a vast and diverse range of BBB modulators. In this review, we summarise and compare the various classes of BBB modulators developed over the last five decades-their recent advancements, advantages and disadvantages, while providing some insight into their future as BBB modulators.
Collapse
Affiliation(s)
- Rory Whelan
- School of Biological and Health Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
- Chemical and Structural Biology, Environmental Sustainability and Health Institute, Technological University Dublin, D07 H6K8 Dublin, Ireland
| | - Grainne C. Hargaden
- School of Chemical and Pharmaceutical Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
| | - Andrew J. S. Knox
- School of Biological and Health Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
- Chemical and Structural Biology, Environmental Sustainability and Health Institute, Technological University Dublin, D07 H6K8 Dublin, Ireland
| |
Collapse
|
23
|
Steeg PS. The blood-tumour barrier in cancer biology and therapy. Nat Rev Clin Oncol 2021; 18:696-714. [PMID: 34253912 DOI: 10.1038/s41571-021-00529-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 02/06/2023]
Abstract
The protective blood-brain barrier has a major role in ensuring normal brain function by severely limiting and tightly controlling the ingress of substances into the brain from the circulation. In primary brain tumours, such as glioblastomas, as well as in brain metastases from cancers in other organs, including lung and breast cancers and melanoma, the blood-brain barrier is modified and is referred to as the blood-tumour barrier (BTB). Alterations in the BTB affect its permeability, and this structure participates in reciprocal regulatory pathways with tumour cells. Importantly, the BTB typically retains a heterogeneous capacity to restrict the penetration of many therapeutic agents into intracranial tumours, and overcoming this challenge is a key to improving the effectiveness of treatment and patient quality of life. Herein, current knowledge of BTB structure and function is reviewed from a cell and cancer biology standpoint, with a focus on findings derived from in vivo models and human tumour specimens. Additionally, how this knowledge can be translated into clinical advances for patients with cancer is discussed.
Collapse
Affiliation(s)
- Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
24
|
Franco R, Rivas-Santisteban R, Navarro G, Reyes-Resina I. Adenosine Receptor Antagonists to Combat Cancer and to Boost Anti-Cancer Chemotherapy and Immunotherapy. Cells 2021; 10:cells10112831. [PMID: 34831054 PMCID: PMC8616521 DOI: 10.3390/cells10112831] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 12/22/2022] Open
Abstract
Extracellular adenosine accumulates in the environment of numerous tumors. For years, this fact has fueled preclinical research to determine whether adenosine receptors (ARs) could be the target to fight cancer. The four ARs discovered so far, A1, A2A, A2B and A3, belong to the class A family of G protein-coupled receptors (GPCRs) and all four have been involved in one way or another in regulating tumor progression. Prompted by the successful anti-cancer immunotherapy, the focus was placed on the ARs more involved in regulation of immune cell differentiation and activation and that are able to establish molecular and functional interactions. This review focuses on the potential of A2A and A2B receptor antagonists in cancer control and in boosting anti-cancer chemotherapy and immunotherapy. The article also overviews the ongoing clinical trials in which A2AR and A2BR ligands are being tested in anti-cancer therapy.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain; (R.R.-S.); (G.N.); (I.R.-R.)
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: or
| | - Rafael Rivas-Santisteban
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain; (R.R.-S.); (G.N.); (I.R.-R.)
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain; (R.R.-S.); (G.N.); (I.R.-R.)
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain; (R.R.-S.); (G.N.); (I.R.-R.)
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
25
|
Hashimoto Y, Campbell M, Tachibana K, Okada Y, Kondoh M. Claudin-5: A Pharmacological Target to Modify the Permeability of the Blood-Brain Barrier. Biol Pharm Bull 2021; 44:1380-1390. [PMID: 34602546 DOI: 10.1248/bpb.b21-00408] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Claudin-5 is the dominant tight junction protein in brain endothelial cells and exclusively limits the paracellular permeability of molecules larger than 400 Da across the blood-brain barrier (BBB). Its pathological impairment or sustained down-regulation has been shown to lead to the progression of psychiatric and neurological disorders, whereas its expression under physiological conditions prevents the passage of drugs across the BBB. While claudin-5 enhancers could potentially act as vascular stabilizers to treat neurological diseases, claudin-5 inhibitors could function as delivery systems to enhance the brain uptake of hydrophilic small-molecular-weight drugs. Therefore, the effects of claudin-5 manipulation on modulating the BBB in different neurological diseases requires further examination. To manipulate claudin-5 expression levels and function, several claudin-5 modulating molecules have been developed. In this review, we first describe the molecular, cellular and pathological aspects of claudin-5 to highlight the mechanisms of claudin-5 enhancers/inhibitors. We then discuss recently developed claudin-5 enhancers/inhibitors and new methods to discover these molecules.
Collapse
Affiliation(s)
| | | | | | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
26
|
Vézina A, Manglani M, Morris D, Foster B, McCord M, Song H, Zhang M, Davis D, Zhang W, Bills J, Nagashima K, Shankarappa P, Kindrick J, Walbridge S, Peer CJ, Figg WD, Gilbert MR, McGavern DB, Muldoon LL, Jackson S. Adenosine A2A Receptor Activation Enhances Blood-Tumor Barrier Permeability in a Rodent Glioma Model. Mol Cancer Res 2021; 19:2081-2095. [PMID: 34521765 DOI: 10.1158/1541-7786.mcr-19-0995] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/16/2020] [Accepted: 09/07/2021] [Indexed: 11/16/2022]
Abstract
The blood-tumor barrier (BTB) limits the entry of effective chemotherapeutic agents into the brain for treatment of malignant tumors like glioblastoma. Poor drug entry across the BTB allows infiltrative glioma stem cells to evade therapy and develop treatment resistance. Regadenoson, an FDA-approved adenosine A2A receptor (A2AR) agonist, has been shown to increase drug delivery across the blood-brain barrier in non-tumor-bearing rodents without a defined mechanism of enhancing BTB permeability. Here, we characterize the time-dependent impact of regadenoson on brain endothelial cell interactions and paracellular transport, using mouse and rat brain endothelial cells and tumor models. In vitro, A2AR activation leads to disorganization of cytoskeletal actin filaments by 30 minutes, downregulation of junctional protein expression by 4 hours, and reestablishment of endothelial cell integrity by 8 hours. In rats bearing intracranial gliomas, regadenoson treatment results in increase of intratumoral temozolomide concentrations, yet no increased survival noted with combined temozolomide therapy. These findings demonstrate regadenoson's ability to induce brain endothelial structural changes among glioma to increase BTB permeability. The use of vasoactive mediators, like regadenoson, which transiently influences paracellular transport, should further be explored to evaluate their potential to enhance central nervous system treatment delivery to aggressive brain tumors. IMPLICATIONS: This study provides insight on the use of a vasoactive agent to increase exposure of the BTB to chemotherapy with intention to improve glioma treatment efficacy.
Collapse
Affiliation(s)
- Amélie Vézina
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland.,Electron Microscope Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Monica Manglani
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - DreeAnna Morris
- Department of Neurology, Oregon Health & Sciences University, Portland, Oregon
| | - Brandon Foster
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | | | - Hua Song
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Meili Zhang
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Dionne Davis
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Wei Zhang
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland
| | - Jessica Bills
- Department of Neurology, Oregon Health & Sciences University, Portland, Oregon
| | - Kunio Nagashima
- Electron Microscope Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Priya Shankarappa
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | - Jessica Kindrick
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | - Stuart Walbridge
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Cody J Peer
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | - William D Figg
- Genitourinary Malignancies Branch, Molecular Pharmacology Section, NCI, NIH, Bethesda, Maryland
| | | | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Leslie L Muldoon
- Department of Neurology, Oregon Health & Sciences University, Portland, Oregon
| | - Sadhana Jackson
- Neuro-Oncology Branch, NCI, NIH, Bethesda, Maryland. .,Electron Microscope Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
27
|
Chowdhury EA, Noorani B, Alqahtani F, Bhalerao A, Raut S, Sivandzade F, Cucullo L. Understanding the brain uptake and permeability of small molecules through the BBB: A technical overview. J Cereb Blood Flow Metab 2021; 41:1797-1820. [PMID: 33444097 PMCID: PMC8327119 DOI: 10.1177/0271678x20985946] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The brain is the most important organ in our body requiring its unique microenvironment. By the virtue of its function, the blood-brain barrier poses a significant hurdle in drug delivery for the treatment of neurological diseases. There are also different theories regarding how molecules are typically effluxed from the brain. In this review, we comprehensively discuss how the different pharmacokinetic techniques used for measuring brain uptake/permeability of small molecules have evolved with time. We also discuss the advantages and disadvantages associated with these different techniques as well as the importance to utilize the right method to properly assess CNS exposure to drug molecules. Even though very strong advances have been made we still have a long way to go to ensure a reduction in failures in central nervous system drug development programs.
Collapse
Affiliation(s)
- Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aditya Bhalerao
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, USA
| | - Snehal Raut
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, USA
| | - Farzane Sivandzade
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, USA
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, USA
| |
Collapse
|
28
|
Schwarzenbach C, Tatsch L, Brandstetter Vilar J, Rasenberger B, Beltzig L, Kaina B, Tomicic MT, Christmann M. Targeting c-IAP1, c-IAP2, and Bcl-2 Eliminates Senescent Glioblastoma Cells Following Temozolomide Treatment. Cancers (Basel) 2021; 13:cancers13143585. [PMID: 34298797 PMCID: PMC8306656 DOI: 10.3390/cancers13143585] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Despite extensive research, malignant glioma remains the most aggressive and fatal type of brain tumor. Following resection, therapy is based on radiation concomitant with the methylating agent temozolomide (TMZ), followed by adjuvant high-dose TMZ. In previous work, we showed that following TMZ exposure, most glioma cells evade apoptosis and enter a senescent state and are thereby protected against anticancer therapy. Senescent cells may escape from senescence, contributing to the formation of recurrences or can induce the senescence-associated secretory phenotype (SASP), which may impact therapy success. Therefore, direct targeting of senescent cells might be favorable to improve the effect of TMZ-based anticancer therapy. Here we show that during TMZ-induced senescence in glioblastoma cells, the antiapoptotic factors c-IAP2 and Bcl-2 are responsible for the prevention of cell death and that inhibition of these factors by BV6 and venetoclax effectively kills senescent glioblastoma cells. Abstract Therapy of malignant glioma depends on the induction of O6-methylguanine by the methylating agent temozolomide (TMZ). However, following TMZ exposure, most glioma cells evade apoptosis and become senescent and are thereby protected against further anticancer therapy. This protection is thought to be dependent on the senescent cell anti-apoptotic pathway (SCAP). Here we analyzed the factors involved in the SCAP upon exposure to TMZ in glioblastoma cell lines (LN-229, A172, U87MG) and examined whether inhibition of these factors could enhance TMZ-based toxicity by targeting senescent cells. We observed that following TMZ treatment, c-IAP2 and Bcl-2 were upregulated. Inhibition of these SCAP factors using non-toxic concentrations of the small molecule inhibitors, BV6 and venetoclax, significantly increased cell death, as measured 144 h after TMZ exposure. Most importantly, BV6 and venetoclax treatment of senescent cells strongly increased cell death after an additional 120 h. Moreover, Combenefit analyses revealed a significant synergy combining BV6 and venetoclax. In contrast to BV6 and venetoclax, AT406, embelin, and TMZ itself, teniposide and the PARP inhibitor pamiparib did not increase cell death in senescent cells. Based on these data, we suggest that BV6 and venetoclax act as senolytic agents in glioblastoma cells upon TMZ exposure.
Collapse
|
29
|
Marcucci F, Corti A, Ferreri AJM. Breaching the Blood-Brain Tumor Barrier for Tumor Therapy. Cancers (Basel) 2021; 13:cancers13102391. [PMID: 34063335 PMCID: PMC8156088 DOI: 10.3390/cancers13102391] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022] Open
Abstract
Tumors affecting the central nervous system (CNS), either primary or secondary, are highly prevalent and represent an unmet medical need. Prognosis of these tumors remains poor, mostly due to the low intrinsic chemo/radio-sensitivity of tumor cells, a meagerly known role of the microenvironment and the poor CNS bioavailability of most used anti-cancer agents. The BBTB is the main obstacle for anticancer drugs to achieve therapeutic concentrations in the tumor tissues. During the last decades, many efforts have been devoted to the identification of modalities allowing to increase drug delivery into brain tumors. Until recently, success has been modest, as few of these approaches reached clinical testing and even less gained regulatory approval. In recent years, the scenario has changed, as various conjugates and drug delivery technologies have advanced into clinical testing, with encouraging results and without being burdened by a heavy adverse event profile. In this article, we review the different approaches aimed at increasing drug delivery to brain tumors, with particular attention to new, promising approaches that increase the permeability of the BBTB or exploit physiological transport mechanisms.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20132 Milan, Italy
- Correspondence: (F.M.); (A.C.)
| | - Angelo Corti
- Division of Experimental Oncology, Tumor Biology and Vascular Targeting Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Correspondence: (F.M.); (A.C.)
| | - Andrés J. M. Ferreri
- Lymphoma Unit, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| |
Collapse
|
30
|
Wala K, Szlasa W, Saczko J, Rudno-Rudzińska J, Kulbacka J. Modulation of Blood-Brain Barrier Permeability by Activating Adenosine A2 Receptors in Oncological Treatment. Biomolecules 2021; 11:biom11050633. [PMID: 33923147 PMCID: PMC8146369 DOI: 10.3390/biom11050633] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
The blood–brain barrier (BBB) plays an important protective role in the central nervous system and maintains its homeostasis. It regulates transport into brain tissue and protects neurons against the toxic effects of substances circulating in the blood. However, in the case of neurological diseases or primary brain tumors, i.e., gliomas, the higher permeability of the blood-derived substances in the brain tissue is necessary. Currently applied methods of treatment for the primary brain neoplasms include surgical removal of the tumor, radiation therapy, and chemotherapy. Despite the abovementioned treatment methods, the prognosis of primary brain tumors remains bad. Moreover, chemotherapy options seem to be limited due to low drug penetration into the cancerous tissue. Modulation of the blood–brain barrier permeability may contribute to an increase in the concentration of the drug in the CNS and thus increase the effectiveness of therapy. Interestingly, endothelial cells in cerebral vessels are characterized by the presence of adenosine 2A receptors (A2AR). It has been shown that substances affecting these receptors regulate the permeability of the BBB. The mechanism of increasing the BBB permeability by A2AR agonists is the actin-cytoskeletal reorganization and acting on the tight junctions. In this case, the A2AR seems to be a promising therapy target. This article aims to assess the possibility of increasing the BBB permeability through A2AR agonists to increase the effectiveness of chemotherapy and to improve the results of cancer therapy.
Collapse
Affiliation(s)
- Kamila Wala
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.W.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.W.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julia Rudno-Rudzińska
- Department of General and Oncological Surgery, Medical University Hospital, Borowska 213, 50-556 Wrocław, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-784-06-92
| |
Collapse
|
31
|
Polak D, Talar M, Wolska N, Wojkowska DW, Karolczak K, Kramkowski K, Bonda TA, Watala C, Przygodzki T. Adenosine Receptor Agonist HE-NECA Enhances Antithrombotic Activities of Cangrelor and Prasugrel in vivo by Decreasing of Fibrinogen Density in Thrombus. Int J Mol Sci 2021; 22:3074. [PMID: 33802928 PMCID: PMC8002731 DOI: 10.3390/ijms22063074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 02/04/2023] Open
Abstract
Blood platelets' adenosine receptors (AR) are considered to be a new target for the anti-platelet therapy. This idea is based on in vitro studies which show that signaling mediated by these receptors leads to a decreased platelet response to activating stimuli. In vivo evidence for the antithrombotic activity of AR agonists published to date were limited, however, to the usage of relatively high doses given in bolus. The present study was aimed at verifying if these substances used in lower doses in combination with inhibitors of P2Y12 could serve as components of dual anti-platelet therapy. We have found that a selective A2A agonist 2-hexynyl-5'-N-ethylcarboxamidoadenosine (HE-NECA) improved the anti-thrombotic properties of either cangrelor or prasugrel in the model of ferric chloride-induced experimental thrombosis in mice. Importantly, HE-NECA was effective not only when applied in bolus as other AR agonists in the up-to-date published studies, but also when given chronically. In vitro thrombus formation under flow conditions revealed that HE-NECA enhanced the ability of P2Y12 inhibitors to decrease fibrinogen content in thrombi, possibly resulting in their lower stability. Adenosine receptor agonists possess a certain hypotensive effect and an ability to increase the blood-brain barrier permeability. Therefore, the effects of anti-thrombotic doses of HE-NECA on blood pressure and the blood-brain barrier permeability in mice were tested. HE-NECA applied in bolus caused a significant hypotension in mice, but the effect was much lower when the substance was given in doses corresponding to that obtained by chronic administration. At the same time, no significant effect of HE-NECA was observed on the blood-brain barrier. We conclude that chronic administration of the A2A agonist can be considered a potential component of a dual antithrombotic therapy. However, due to the hypotensive effect of the substances, dosage and administration must be elaborated to minimize the side-effects. The total number of animals used in the experiments was 146.
Collapse
Affiliation(s)
- Dawid Polak
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| | - Marcin Talar
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| | - Nina Wolska
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| | - Dagmara W. Wojkowska
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| | - Kamil Karolczak
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| | - Karol Kramkowski
- Department of Physical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-089 Bialystok, Poland;
| | - Tomasz A. Bonda
- Department of General and Experimental Pathology, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland;
| | - Cezary Watala
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| | - Tomasz Przygodzki
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235 Lodz, Poland; (D.P.); (M.T.); (N.W.); (D.W.W.); (K.K.); (C.W.)
| |
Collapse
|
32
|
Strategies for delivering therapeutics across the blood-brain barrier. Nat Rev Drug Discov 2021; 20:362-383. [PMID: 33649582 DOI: 10.1038/s41573-021-00139-y] [Citation(s) in RCA: 424] [Impact Index Per Article: 141.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2021] [Indexed: 02/06/2023]
Abstract
Achieving sufficient delivery across the blood-brain barrier is a key challenge in the development of drugs to treat central nervous system (CNS) disorders. This is particularly the case for biopharmaceuticals such as monoclonal antibodies and enzyme replacement therapies, which are largely excluded from the brain following systemic administration. In recent years, increasing research efforts by pharmaceutical and biotechnology companies, academic institutions and public-private consortia have resulted in the evaluation of various technologies developed to deliver therapeutics to the CNS, some of which have entered clinical testing. Here we review recent developments and challenges related to selected blood-brain barrier-crossing strategies - with a focus on non-invasive approaches such as receptor-mediated transcytosis and the use of neurotropic viruses, nanoparticles and exosomes - and analyse their potential in the treatment of CNS disorders.
Collapse
|
33
|
Ghiaseddin A, Hoang Minh LB, Janiszewska M, Shin D, Wick W, Mitchell DA, Wen PY, Grossman SA. Adult precision medicine: learning from the past to enhance the future. Neurooncol Adv 2021; 3:vdaa145. [PMID: 33543142 PMCID: PMC7846182 DOI: 10.1093/noajnl/vdaa145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite therapeutic advances for other malignancies, gliomas remain challenging solid tumors to treat. Complete surgical resection is nearly impossible due to gliomas’ diffuse infiltrative nature, and treatment is hampered by restricted access to the tumors due to limited transport across the blood–brain barrier. Recent advances in genomic studies and next-generation sequencing techniques have led to a better understanding of gliomas and identification of potential aberrant signaling pathways. Targeting the specific genomic abnormalities via novel molecular therapies has opened a new avenue in the management of gliomas, with encouraging results in preclinical studies and early clinical trials. However, molecular characterization of gliomas revealed significant heterogeneity, which poses a challenge for targeted therapeutic approaches. In this context, leading neuro-oncology researchers and clinicians, industry innovators, and patient advocates convened at the inaugural annual Remission Summit held in Orlando, FL in February 2019 to discuss the latest advances in immunotherapy and precision medicine approaches for the treatment of adult and pediatric brain tumors and outline the unanswered questions, challenges, and opportunities that lay ahead for advancing the duration and quality of life for patients with brain tumors. Here, we provide historical context for precision medicine in other cancers, present emerging approaches for gliomas, discuss their limitations, and outline the steps necessary for future success. We focus on the advances in small molecule targeted therapy, as the use of immunotherapy as an emerging precision medicine modality for glioma treatment has recently been reviewed by our colleagues.
Collapse
Affiliation(s)
- Ashley Ghiaseddin
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Lan B Hoang Minh
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | | | - David Shin
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Wolfgang Wick
- Neurology Clinic, Heidelberg University Medical Center, Heidelberg, Germany
| | - Duane A Mitchell
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Patrick Y Wen
- Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA.,Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Stuart A Grossman
- Department of Oncology, Johns Hopkins Kimmel Cancer Center, Baltimore, Maryland, USA
| |
Collapse
|
34
|
Abstract
Cancer treatment remains a challenge due to a high level of intra- and intertumoral heterogeneity and the rapid development of chemoresistance. In the brain, this is further hampered by the blood-brain barrier that reduces passive diffusion of drugs to a minimum. Tumors grow invasively and form new blood vessels, also in brain tissue where remodeling of pre-existing vasculature is substantial. The cancer-associated vessels in the brain are considered leaky and thus could facilitate the transport of chemotherapeutic agents. Yet, brain tumors are extremely difficult to treat, and, in this review, we will address how different aspects of the vasculature in brain tumors contribute to this.
Collapse
Affiliation(s)
- Casper Hempel
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark.
| | - Kasper B Johnsen
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark
| | - Serhii Kostrikov
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark
| | - Petra Hamerlik
- Brain Tumor Biology, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Thomas L Andresen
- Dept of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark.
| |
Collapse
|
35
|
Central nervous system delivery of molecules across the blood-brain barrier. Neurochem Int 2021; 144:104952. [PMID: 33400964 DOI: 10.1016/j.neuint.2020.104952] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/15/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022]
Abstract
Therapies targeting neurological conditions such as Alzheimer's or Parkinson's diseases are hampered by the presence of the blood-brain barrier (BBB). During the last decades, several approaches have been developed to overcome the BBB, such as the use of nanoparticles (NPs) based on biomaterials, or alternative methods to open the BBB. In this review, we briefly highlight these strategies and the most recent advances in this field. Limitations and advantages of each approach are discussed. Combination of several methods such as functionalized NPs targeting the receptor-mediated transcytosis system with the use of magnetic resonance imaging-guided focused ultrasound (FUS) might be a promising strategy to develop theranostic tools as well as to safely deliver therapeutic molecules, such as drugs, neurotrophic factors or antibodies within the brain parenchyma.
Collapse
|
36
|
Blood-Brain Barrier Modulation to Improve Glioma Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12111085. [PMID: 33198244 PMCID: PMC7697580 DOI: 10.3390/pharmaceutics12111085] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by brain microvascular endothelial cells that are sealed by tight junctions, making it a significant obstacle for most brain therapeutics. The poor BBB penetration of newly developed therapeutics has therefore played a major role in limiting their clinical success. A particularly challenging therapeutic target is glioma, which is the most frequently occurring malignant brain tumor. Thus, to enhance therapeutic uptake in tumors, researchers have been developing strategies to modulate BBB permeability. However, most conventional BBB opening strategies are difficult to apply in the clinical setting due to their broad, non-specific modulation of the BBB, which can result in damage to normal brain tissue. In this review, we have summarized strategies that could potentially be used to selectively and efficiently modulate the tumor BBB for more effective glioma treatment.
Collapse
|
37
|
Zhang D, Wang Y, Lin H, Sun Y, Wang M, Jia Y, Yu X, Jiang H, Xu W, Sun JP, Xu Z. Function and therapeutic potential of G protein-coupled receptors in epididymis. Br J Pharmacol 2020; 177:5489-5508. [PMID: 32901914 DOI: 10.1111/bph.15252] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/08/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022] Open
Abstract
Infertility rates for both females and males have increased continuously in recent years. Currently, effective treatments for male infertility with defined mechanisms or targets are still lacking. G protein-coupled receptors (GPCRs) are the largest class of drug targets, but their functions and the implications for the therapeutic development for male infertility largely remain elusive. Nevertheless, recent studies have shown that several members of the GPCR superfamily play crucial roles in the maintenance of ion-water homeostasis of the epididymis, development of the efferent ductules, formation of the blood-epididymal barrier and maturation of sperm. Knowledge of the functions, genetic variations and working mechanisms of such GPCRs, along with the drugs and ligands relevant to their specific functions, provide future directions and a great arsenal for new developments in the treatment of male infertility.
Collapse
Affiliation(s)
- Daolai Zhang
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China.,Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Yanfei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Hui Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Yujing Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Mingwei Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Yingli Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Xiao Yu
- Department of Physiology, School of Medicine, Shandong University, Jinan, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing, China
| | - Wenming Xu
- Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University West China Second University Hospital, Chengdu, China
| | - Jin-Peng Sun
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China.,Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, China
| |
Collapse
|
38
|
Karmur BS, Philteos J, Abbasian A, Zacharia BE, Lipsman N, Levin V, Grossman S, Mansouri A. Blood-Brain Barrier Disruption in Neuro-Oncology: Strategies, Failures, and Challenges to Overcome. Front Oncol 2020; 10:563840. [PMID: 33072591 PMCID: PMC7531249 DOI: 10.3389/fonc.2020.563840] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/13/2020] [Indexed: 01/05/2023] Open
Abstract
The blood-brain barrier (BBB) presents a formidable challenge in the development of effective therapeutics in neuro-oncology. This has fueled several decades of efforts to develop strategies for disrupting the BBB, but progress has not been satisfactory. As such, numerous drug- and device-based methods are currently being investigated in humans. Through a focused assessment of completed, active, and pending clinical trials, our first aim in this review is to outline the scientific foundation, successes, and limitations of the BBBD strategies developed to date. Among 35 registered trials relevant to BBBD in neuro-oncology in the ClinicalTrials.gov database, mannitol was the most common drug-based method, followed by RMP-7 and regadenoson. MR-guided focused ultrasound was the most common device-based method, followed by MR-guided laser ablation, ultrasound, and transcranial magnetic stimulation. While most early-phase studies focusing on safety and tolerability have met stated objectives, advanced-phase studies focusing on survival differences and objective tumor response have been limited by heterogeneous populations and tumors, along with a lack of control arms. Based on shared challenges among all methods, our second objective is to discuss strategies for confirmation of BBBD, choice of systemic agent and drug design, alignment of BBBD method with real-world clinical workflow, and consideration of inadvertent toxicity associated with disrupting an evolutionarily-refined barrier. Finally, we conclude with a strategic proposal to approach future studies assessing BBBD.
Collapse
Affiliation(s)
- Brij S Karmur
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Aram Abbasian
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Brad E Zacharia
- Penn State Health Neurosurgery, College of Medicine, Penn State University, Hershey, PA, United States
| | - Nir Lipsman
- Division of Neurosurgery, University of Toronto, Toronto, ON, Canada
| | - Victor Levin
- Department of Neurosurgery, Medical School, University of California, San Francisco, San Francisco, CA, United States
| | - Stuart Grossman
- Department of Oncology, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Alireza Mansouri
- Penn State Health Neurosurgery, College of Medicine, Penn State University, Hershey, PA, United States
| |
Collapse
|
39
|
Pierce CF, Kwasnicki A, Lakka SS, Engelhard HH. Cerebral Microdialysis as a Tool for Assessing the Delivery of Chemotherapy in Brain Tumor Patients. World Neurosurg 2020; 145:187-196. [PMID: 32890850 DOI: 10.1016/j.wneu.2020.08.161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/27/2022]
Abstract
The development of curative treatment for glioblastoma has been extremely challenging. Chemotherapeutic agents that have seemed promising have failed in clinical trials. Drugs that can successfully target cancer cells within the brain must first traverse the brain interstitial fluid. Cerebral microdialysis (CMD) is an invasive technique in which interstitial fluid can be directly sampled. CMD has primarily been used clinically in the setting of head trauma and subarachnoid hemorrhage. Our goal was to review the techniques, principles, and new data pertaining to CMD to highlight its use in neuro-oncology. We conducted a literature search using the PubMed database and selected studies in which the investigators had used CMD in either animal brain tumor models or clinical trials. The references were reviewed for additional information. Studies of CMD have shown its importance as a neurosurgical technique. CMD allows for the collection of pharmacokinetic data on drug penetrance across the blood-brain barrier and metabolic data to characterize the response to chemotherapy. Although no complications have been reported, the current CMD technique (as with any procedure) has risks and limitations, which we have described in the present report. Animal CMD experiments have been used to exclude central nervous system drug candidates from progressing to clinical trials. At present, patients undergoing CMD have been monitored in the intensive care unit, owing to the requisite tethering to the apparatus. This can be expected to change soon because of advances in microminiaturization. CMD is an extremely valuable, yet underused, technique. Future CMD applications will have central importance in assessing drug delivery to tumor cells in vivo, allowing a pathway to successful therapy for malignant brain tumors.
Collapse
Affiliation(s)
- Charles F Pierce
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amanda Kwasnicki
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sajani S Lakka
- Department of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Herbert H Engelhard
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA; Department of Bioengineering, The University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
40
|
Herbener VJ, Burster T, Goreth A, Pruss M, von Bandemer H, Baisch T, Fitzel R, Siegelin MD, Karpel-Massler G, Debatin KM, Westhoff MA, Strobel H. Considering the Experimental use of Temozolomide in Glioblastoma Research. Biomedicines 2020; 8:E151. [PMID: 32512726 PMCID: PMC7344626 DOI: 10.3390/biomedicines8060151] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/30/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Temozolomide (TMZ) currently remains the only chemotherapeutic component in the approved treatment scheme for Glioblastoma (GB), the most common primary brain tumour with a dismal patient's survival prognosis of only ~15 months. While frequently described as an alkylating agent that causes DNA damage and thus-ultimately-cell death, a recent debate has been initiated to re-evaluate the therapeutic role of TMZ in GB. Here, we discuss the experimental use of TMZ and highlight how it differs from its clinical role. Four areas could be identified in which the experimental data is particularly limited in its translational potential: 1. transferring clinical dosing and scheduling to an experimental system and vice versa; 2. the different use of (non-inert) solvent in clinic and laboratory; 3. the limitations of established GB cell lines which only poorly mimic GB tumours; and 4. the limitations of animal models lacking an immune response. Discussing these limitations in a broader biomedical context, we offer suggestions as to how to improve transferability of data. Finally, we highlight an underexplored function of TMZ in modulating the immune system, as an example of where the aforementioned limitations impede the progression of our knowledge.
Collapse
Affiliation(s)
- Verena J. Herbener
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Alicia Goreth
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Maximilian Pruss
- Department of Gynecology and Obstetrics, Medical Faculty, University Hospital of the Heinrich-Heine-University Duesseldorf, D-40225 Duesseldorf, Germany;
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Hélène von Bandemer
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Tim Baisch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Rahel Fitzel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Markus D. Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, D-89081 Ulm, Germany;
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany; (V.J.H.); (A.G.); (H.v.B.); (T.B.); (R.F.); (K.-M.D.); (H.S.)
| |
Collapse
|
41
|
Lynes JP, Nwankwo AK, Sur HP, Sanchez VE, Sarpong KA, Ariyo OI, Dominah GA, Nduom EK. Biomarkers for immunotherapy for treatment of glioblastoma. J Immunother Cancer 2020; 8:e000348. [PMID: 32474411 PMCID: PMC7264836 DOI: 10.1136/jitc-2019-000348] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2020] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy is a promising new therapeutic field that has demonstrated significant benefits in many solid-tumor malignancies, such as metastatic melanoma and non-small cell lung cancer. However, only a subset of these patients responds to treatment. Glioblastoma (GBM) is the most common malignant primary brain tumor with a poor prognosis of 14.6 months and few treatment advancements over the last 10 years. There are many clinical trials testing immune therapies in GBM, but patient responses in these studies have been highly variable and a definitive benefit has yet to be identified. Biomarkers are used to quantify normal physiology and physiological response to therapies. When extensively characterized and vigorously validated, they have the potential to delineate responders from non-responders for patients treated with immunotherapy in malignancies outside of the central nervous system (CNS) as well as GBM. Due to the challenges of current modalities of radiographic diagnosis and disease monitoring, identification of new predictive and prognostic biomarkers to gauge response to immune therapy for patients with GBM will be critical in the precise treatment of this highly heterogenous disease. This review will explore the current and future strategies for the identification of potential biomarkers in the field of immunotherapy for GBM, as well as highlight major challenges of adapting immune therapy for CNS malignancies.
Collapse
Affiliation(s)
- John P Lynes
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Anthony K Nwankwo
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Hannah P Sur
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Victoria E Sanchez
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Kwadwo A Sarpong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Oluwatobi I Ariyo
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Gifty A Dominah
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Edjah K Nduom
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
42
|
Zyma M, Pawliczak R. Characteristics and the role of purinergic receptors in pathophysiology with focus on immune response. Int Rev Immunol 2020; 39:97-117. [PMID: 32037918 DOI: 10.1080/08830185.2020.1723582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The nucleotide adenosine-5'-triphosphate (ATP) is mostly thought to be energy carrier, but evidence presented in multiple studies proves ATP involvement into variety of processes, due to its neuromodulatory capabilities. ATP and its metabolite-adenosine, bind to the purinergic receptors, which are divided into two types: adenosine binding P1 receptor and ADP/ATP binding P2 receptor. These receptors are expressed in different tissues and organs. Recent studies report their immunomodulatory characteristics, connected with varying immunological processes, such as immunological response or antigen presentation. Besides, they seem to play an important role in medical conditions such as bronchial asthma or variety of cancers. In this article, we would like to review recent discoveries on the field of purinergic receptors research focusing on their role in immunological system, and shed a new light upon the importance of these receptors in modern medicine development.
Collapse
Affiliation(s)
- Marharyta Zyma
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
43
|
Li Z, Li F, Ma C, Xu C, Pan Z. Advancement of clinical therapeutic research on glioma: A narrative review. GLIOMA 2020. [DOI: 10.4103/glioma.glioma_18_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
44
|
Adenosine Receptor Agonists Exhibit Anti-Platelet Effects and the Potential to Overcome Resistance to P2Y 12 Receptor Antagonists. Molecules 2019; 25:molecules25010130. [PMID: 31905703 PMCID: PMC6982709 DOI: 10.3390/molecules25010130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/16/2019] [Accepted: 12/25/2019] [Indexed: 11/30/2022] Open
Abstract
Large inter-individual variation in platelet response to endogenous agonists and pharmacological agents, including resistance to antiplatelet therapy, prompts a search for novel platelet inhibitors and development new antithrombotic strategies. The present in vitro study evaluates the beneficial effects of three adenosine receptor (AR) agonists (regadenoson, LUF 5835 and NECA), different in terms of their selectivity for platelet adenosine receptors, when used alone and in combination with P2Y12 inhibitors, such as cangrelor or prasugrel metabolite. The anti-platelet effects of AR agonists were evaluated in healthy subjects (in the whole group and after stratification of individuals into high- and low-responders to P2Y12 inhibitors), using whole blood techniques, under flow (thrombus formation) and static conditions (study of platelet activation and aggregation). Compared to P2Y12 antagonists, AR agonists were much less or not effective under static conditions, but demonstrated similar antiplatelet activity in flow. In most cases, AR agonists significantly enhanced the anti-platelet effect of P2Y12 antagonists, despite possessing different selectivity profiles and antiplatelet activities. Importantly, their inhibitory effects in combination with P2Y12 antagonists were similar in high- and low-responders to P2Y12 inhibitors. In conclusion, a combination of anti-platelet agents acting via the P1 and P2 purinergic receptors represents a promising alternative to existing antithrombotic therapy.
Collapse
|
45
|
Stepanenko AA, Chekhonin VP. On the Critical Issues in Temozolomide Research in Glioblastoma: Clinically Relevant Concentrations and MGMT-independent Resistance. Biomedicines 2019; 7:biomedicines7040092. [PMID: 31783653 PMCID: PMC6966644 DOI: 10.3390/biomedicines7040092] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
The current standard first-line treatment for adult patients with newly diagnosed glioblastoma includes concurrent radiotherapy and daily oral temozolomide (TMZ), followed by adjuvant TMZ. As a prodrug, TMZ undergoes spontaneous hydrolysis generating a methylating agent. O6-methylguanine is considered the most preponderant toxic damage mechanism at therapeutically relevant TMZ doses, whereas MGMT, which encodes the O6-methylguanine-DNA methyltransferase DNA repair enzyme, is the most relevant resistance mechanism. Speculations on clinically relevant TMZ concentrations, cytotoxic and cytostatic effects of TMZ, and resistance mechanisms exist in the literature. Here, we raise the following principal issues: What are the clinically relevant TMZ concentrations in glioma patients, and which TMZ-induced molecular lesion(s) and corresponding resistance mechanism(s) are important for TMZ therapeutic effects at clinically relevant concentrations? According to clinical data from patients with glioblastoma, the mean peak TMZ concentrations in the peritumoral tissue might be much lower (around 5 µM) than usually used in in vitro research, and may represent only 20% of systemic drug levels. According to in vitro reports, single-dose TMZ at concentrations around 5 µM have minimal, if any, effect on apoptosis and/or senescence of glioblastoma cell lines. However, the clinically relevant concentrations of TMZ are sufficient to radiosensitize both MGMT-positive and -negative cell lines in vitro. It is speculated that a single DNA repair protein, MGMT, is highly efficient in protecting cells against TMZ toxicity. However, an endogenous level of MGMT protein expression is not universally correlated with TMZ responsiveness, and MGMT-independent mechanisms of TMZ resistance exist.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia;
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov Str. 1, 117997 Moscow, Russia
- Correspondence:
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Kropotkinsky Lane 23, 119034 Moscow, Russia;
- Department of Medical Nanobiotechnologies, Medico-Biological Faculty, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Ostrovitianov Str. 1, 117997 Moscow, Russia
| |
Collapse
|
46
|
Fisher DG, Price RJ. Recent Advances in the Use of Focused Ultrasound for Magnetic Resonance Image-Guided Therapeutic Nanoparticle Delivery to the Central Nervous System. Front Pharmacol 2019; 10:1348. [PMID: 31798453 PMCID: PMC6864822 DOI: 10.3389/fphar.2019.01348] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Targeting systemically-administered drugs and genes to specific regions of the central nervous system (CNS) remains a challenge. With applications extending into numerous disorders and cancers, there is an obvious need for approaches that facilitate the delivery of therapeutics across the impervious blood-brain barrier (BBB). Focused ultrasound (FUS) is an emerging treatment method that leverages acoustic energy to oscillate simultaneously administered contrast agent microbubbles. This FUS-mediated technique temporarily disrupts the BBB, allowing ordinarily impenetrable agents to diffuse and/or convect into the CNS. Under magnetic resonance image guidance, FUS and microbubbles enable regional targeting-limiting the large, and potentially toxic, dosage that is often characteristic of systemically-administered therapies. Subsequent to delivery across the BBB, therapeutics face yet another challenge: penetrating the electrostatically-charged, mesh-like brain parenchyma. Non-bioadhesive, encapsulated nanoparticles can help overcome this additional barrier to promote widespread treatment in selected target areas. Furthermore, nanoparticles offer significant advantages over conventional systemically-administered therapeutics. Surface modifications of nanoparticles can be engineered to enhance targeted cellular uptake, and nanoparticle formulations can be tailored to control many pharmacokinetic properties such as rate of drug liberation, distribution, and excretion. For instance, nanoparticles loaded with gene plasmids foster relatively stable transfection, thus obviating the need for multiple, successive treatments. As the formulations and applications of these nanoparticles can vary greatly, this review article provides an overview of FUS coupled with polymeric or lipid-based nanoparticles currently utilized for drug delivery, diagnosis, and assessment of function in the CNS.
Collapse
Affiliation(s)
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
47
|
Patties I, Kallendrusch S, Böhme L, Kendzia E, Oppermann H, Gaunitz F, Kortmann RD, Glasow A. The Chk1 inhibitor SAR-020106 sensitizes human glioblastoma cells to irradiation, to temozolomide, and to decitabine treatment. J Exp Clin Cancer Res 2019; 38:420. [PMID: 31639020 PMCID: PMC6805470 DOI: 10.1186/s13046-019-1434-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022] Open
Abstract
Background Glioblastoma is the most common and aggressive brain tumour in adults with a median overall survival of only 14 months after standard therapy with radiation therapy (IR) and temozolomide (TMZ). In a novel multimodal treatment approach we combined the checkpoint kinase 1 (Chk1) inhibitor SAR-020106 (SAR), disrupting homologue recombination, with standard DNA damage inducers (IR, TMZ) and the epigenetic/cytotoxic drug decitabine (5-aza-2′-deoxycitidine, 5-aza-dC). Different in vitro glioblastoma models are monitored to evaluate if the impaired DNA damage repair may chemo/radiosensitize the tumour cells. Methods Human p53-mutated (p53-mut) and -wildtype (p53-wt) glioblastoma cell lines (p53-mut: LN405, T98G; p53-wt: A172, DBTRG) and primary glioblastoma cells (p53-mut: P0297; p53-wt: P0306) were treated with SAR combined with TMZ, 5-aza-dC, and/or IR and analysed for induction of apoptosis (AnnexinV and sub-G1 assay), cell cycle distribution (nuclear PI staining), DNA damage (alkaline comet or gH2A.X assay), proliferation inhibition (BrdU assay), reproductive survival (clonogenic assay), and potential tumour stem cells (nestinpos/GFAPneg fluorescence staining). Potential treatment-induced neurotoxicity was evaluated on nestin-positive neural progenitor cells in a murine entorhinal-hippocampal slice culture model. Results SAR showed radiosensitizing effects on the induction of apoptosis and on the reduction of long-term survival in p53-mut and p53-wt glioblastoma cell lines and primary cells. In p53-mut cells, this effect was accompanied by an abrogation of the IR-induced G2/M arrest and an enhancement of IR-induced DNA damage by SAR treatment. Also TMZ and 5-aza-dC acted radioadditively albeit to a lesser extent. The multimodal treatment achieved the most effective reduction of clonogenicity in all tested cell lines and did not affect the ratio of nestinpos/GFAPneg cells. No neurotoxic effects were detected when the number of nestin-positive neural progenitor cells remained unchanged after multimodal treatment. Conclusion The Chk1 inhibitor SAR-020106 is a potent sensitizer for DNA damage-induced cell death in glioblastoma therapy strongly reducing clonogenicity of tumour cells. Selectively enhanced p53-mut cell death may provide stronger responses in tumours defective of non-homologous end joining (NHEJ). Our results suggest that a multimodal therapy involving DNA damage inducers and DNA repair inhibitors might be an effective anti-tumour strategy with a low risk of neurotoxicity.
Collapse
|
48
|
Li Y, Chen F, Chu J, Wu C, Li Y, Li H, Ma H. miR-148-3p Inhibits Growth of Glioblastoma Targeting DNA Methyltransferase-1 (DNMT1). Oncol Res 2019; 27:911-921. [PMID: 30982493 PMCID: PMC7848282 DOI: 10.3727/096504019x15516966905337] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To date, miR-148-3p and DNMT1–recombinant human runt-related transcription factor 3 (RUNX3) axis have been linked to cell proliferation, migration, and invasion; however, their roles and relationships in human glioblastoma multiforme (GBM) are still not clear. Here we found that the expression of miR-148-3p in glioma tissues was decreased compared with adjacent nontumor tissues and correlated with WHO grade, tumor size, and prognosis as well as DNMT1 and RUNX3 expressions. Compared with NHA cells, the expression of miR-148-3p in U87 and U251 cells was also downregulated and accompanied with upregulation of DNMT1 and hypermethylation level of RUNX3 promoter region. miR-148-3p overexpression induced apoptosis and cell cycle arrest of U87 and U251 cells, and affected cell migration and invasion. miR-148-3p mimics effectively suppressed the expression of DNMT1 and methylation of RUNX3 promoter, finally upregulating RUNX3 expression. Mechanistically, the 3′-untranslated region (3′-UTR) of DNMT1 was a direct target of miR-148-3p. Overexpression of miR-148-3p or inhibition of DNMT1 induced the expression of E-cadherin and reduced the expressions of N-cadherin, vimentin, MMP-2, and MMP-9. In conclusion, miR-148-3p directly repressed the expression of DNMT1 and inhibited proliferation, migration, and invasion by regulating DNMT1–RUNX3 axis and the epithelial–mesenchymal transition in GBM. Our findings provide a new foundation for treatment of patients with GBM.
Collapse
Affiliation(s)
- Yongtao Li
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Fanyu Chen
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Jiancheng Chu
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Chao Wu
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Yuan Li
- School of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Heng Li
- School of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Hongxin Ma
- Department of Neurosurgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| |
Collapse
|
49
|
Jacobson KA, Tosh DK, Jain S, Gao ZG. Historical and Current Adenosine Receptor Agonists in Preclinical and Clinical Development. Front Cell Neurosci 2019; 13:124. [PMID: 30983976 PMCID: PMC6447611 DOI: 10.3389/fncel.2019.00124] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
Adenosine receptors (ARs) function in the body’s response to conditions of pathology and stress associated with a functional imbalance, such as in the supply and demand of energy/oxygen/nutrients. Extracellular adenosine concentrations vary widely to raise or lower the basal activation of four subtypes of ARs. Endogenous adenosine can correct an energy imbalance during hypoxia and other stress, for example, by slowing the heart rate by A1AR activation or increasing the blood supply to heart muscle by the A2AAR. Moreover, exogenous AR agonists, antagonists, or allosteric modulators can be applied for therapeutic benefit, and medicinal chemists working toward that goal have reported thousands of such agents. Thus, numerous clinical trials have ensued, using promising agents to modulate adenosinergic signaling, most of which have not succeeded. Currently, short-acting, parenteral agonists, adenosine and Regadenoson, are the only AR agonists approved for human use. However, new concepts and compounds are currently being developed and applied toward preclinical and clinical evaluation, and initial results are encouraging. This review focuses on key compounds as AR agonists and positive allosteric modulators (PAMs) for disease treatment or diagnosis. AR agonists for treating inflammation, pain, cancer, non-alcoholic steatohepatitis, angina, sickle cell disease, ischemic conditions and diabetes have been under development. Multiple clinical trials with two A3AR agonists are ongoing.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
50
|
Umlauf BJ, Shusta EV. Exploiting BBB disruption for the delivery of nanocarriers to the diseased CNS. Curr Opin Biotechnol 2019; 60:146-152. [PMID: 30849699 DOI: 10.1016/j.copbio.2019.01.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/20/2018] [Accepted: 01/21/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Benjamin J Umlauf
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, United States
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, United States.
| |
Collapse
|