1
|
Werner MS, Aras S, Morgan AR, Roamer J, Param NJ, Olagbegi K, Lamontagne RJ, Greig JA, Wilson JM. Adeno-associated virus-mediated trastuzumab delivery to the central nervous system for human epidermal growth factor receptor 2+ brain metastasis. Cancer Gene Ther 2024; 31:766-777. [PMID: 38480976 DOI: 10.1038/s41417-024-00751-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 05/19/2024]
Abstract
Trastuzumab improves overall survival for HER2+ breast cancer, but its short half-life in the cerebrospinal fluid (~2-4 days) and delivery limitations restrict the ability to target HER2+ central nervous system (CNS) disease. We developed an adeno-associated virus (AAV) vector expressing a codon-optimized, ubiquitin C (UbC)-promoter-driven trastuzumab sequence (AAV9.UbC.trastuzumab) for intrathecal administration. Transgene expression was evaluated in adult Rag1 knockout mice and rhesus nonhuman primates (NHPs) after a single intracerebroventricular (ICV) or intra-cisterna magna (ICM) AAV9.UbC.trastuzumab injection, respectively, using real-time PCR, ELISA, Western blot, in situ hybridization, single-nucleus RNA sequencing, and liquid chromatography-mass spectrometry; antitumor efficacy was evaluated in brain xenografts using HER2+ breast cancer cell lines (BT-474, MDA-MB-453). Transgene expression was detected in brain homogenates of Rag1 knockout mice following a single ICV injection of AAV9.UbC.trastuzumab (1 × 1011 vector genome copies [GC]/mouse) and tumor progression was inhibited in xenograft models of breast-to-brain metastasis. In NHPs, ICM delivery of AAV9.UbC.trastuzumab (3 × 1013 GC/animal) was well tolerated (36-37 days in-life) and resulted in transgene expression in CNS tissues and cerebrospinal fluid at levels sufficient to induce complete tumor remission in MDA-MB-453 brain xenografts. With AAV9's proven clinical safety record, this gene therapy may represent a viable approach for targeting HER2 + CNS malignancies.
Collapse
Affiliation(s)
- Marcela S Werner
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shweta Aras
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ashleigh R Morgan
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jillian Roamer
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nesteene J Param
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kanyin Olagbegi
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Jason Lamontagne
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Holdaway M, Ablyazova F, Huda S, D'Amico RS, Wong T, Shani D, Ben-Shalom N, Boockvar JA. First in-human intrathecal delivery of bevacizumab for leptomeningeal spread from recurrent glioblastoma: rationale for a dose escalation trial. J Neurooncol 2023; 164:231-237. [PMID: 37548850 DOI: 10.1007/s11060-023-04412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/28/2023] [Indexed: 08/08/2023]
Abstract
PURPOSE To outline the dose rationale for the first in-human intrathecal delivery of bevacizumab for LMS of GBM. METHODS A 19-year-old female patient presented to Lenox Hill Hospital following thalamic GBM recurrence. She subsequently underwent two infusions of intra-arterial BEV (NCT01269853) and experienced a period of relative disease stability until progression in 2022. One month later, MRI disclosed diffuse enhancement representative of LMS of GBM. The patient subsequently underwent five cycles of IT BEV in mid-2022 (IND 162119). Doses of 25 mg, 37.5 mg, 50 mg, 50 mg, and 37.8 mg were delivered at two-week intervals between doses 1-4. The final 37.8 mg dose was given one day following her fourth dose, given that the patient was to be discharged, traveled several hours to our center, and was tolerating therapy well. Dosage was decreased due to the short interval between the final two treatments. Shortly after IT BEV completion, she received a third dose of IA BEV. RESULTS Our patient did not show any signs of serious adverse effects or dose limiting toxicities following any of the treatments. It is difficult to determine PFS due to the rapid progression associated with LMS of GBM and rapid timeframe of treatment. CONCLUSION LMS continues to be a devastating progression in many types of cancer, including GBM, and novel ways to deliver therapeutics may offer patients symptomatic and therapeutic benefits.
Collapse
Affiliation(s)
- Matthew Holdaway
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Lenox Hill Hospital, 130 East 77th Street, New York, NY, 10065, USA.
- Albany Medical College, Albany, NY, USA.
| | - Faina Ablyazova
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Lenox Hill Hospital, 130 East 77th Street, New York, NY, 10065, USA
| | - Shayan Huda
- CUNY School of Medicine, New York City, NY, USA
| | - Randy S D'Amico
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Lenox Hill Hospital, 130 East 77th Street, New York, NY, 10065, USA
| | - Tamika Wong
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Lenox Hill Hospital, 130 East 77th Street, New York, NY, 10065, USA
| | - Dana Shani
- Department of Hematology, Medical Oncology, Internal Medicine, Lenox Hill Hospital, New York City, NY, USA
- Northwell Health Cancer Institute, New Hyde Park, New York, NY, USA
| | - Netanel Ben-Shalom
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Lenox Hill Hospital, 130 East 77th Street, New York, NY, 10065, USA
| | - John A Boockvar
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Lenox Hill Hospital, 130 East 77th Street, New York, NY, 10065, USA
| |
Collapse
|
3
|
Grabowski T, Leuschner J, Gad S. 4-Week toxicity study of biosimilar natalizumab in comparison to Tysabri ® by repeated intravenous infusion to cynomolgus monkeys. Drug Chem Toxicol 2022; 45:499-506. [DOI: 10.1080/01480545.2020.1722155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
| | - Jost Leuschner
- LPT Laboratory of Pharmacology and Toxicology GmbH & Co. KG, Hamburg, Germany
| | - Shayne Gad
- Gad Consulting Services, Raleigh, NC, USA
| |
Collapse
|
4
|
Naseri Kouzehgarani G, Feldsien T, Engelhard HH, Mirakhur KK, Phipps C, Nimmrich V, Clausznitzer D, Lefebvre DR. Harnessing cerebrospinal fluid circulation for drug delivery to brain tissues. Adv Drug Deliv Rev 2021; 173:20-59. [PMID: 33705875 DOI: 10.1016/j.addr.2021.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 12/31/2022]
Abstract
Initially thought to be useful only to reach tissues in the immediate vicinity of the CSF circulatory system, CSF circulation is now increasingly viewed as a viable pathway to deliver certain therapeutics deeper into brain tissues. There is emerging evidence that this goal is achievable in the case of large therapeutic proteins, provided conditions are met that are described herein. We show how fluid dynamic modeling helps predict infusion rate and duration to overcome high CSF turnover. We posit that despite model limitations and controversies, fluid dynamic models, pharmacokinetic models, preclinical testing, and a qualitative understanding of the glymphatic system circulation can be used to estimate drug penetration in brain tissues. Lastly, in addition to highlighting landmark scientific and medical literature, we provide practical advice on formulation development, device selection, and pharmacokinetic modeling. Our review of clinical studies suggests a growing interest for intra-CSF delivery, particularly for targeted proteins.
Collapse
|
5
|
Chang HY, Wu S, Meno-Tetang G, Shah DK. A translational platform PBPK model for antibody disposition in the brain. J Pharmacokinet Pharmacodyn 2019; 46:319-338. [PMID: 31115858 DOI: 10.1007/s10928-019-09641-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022]
Abstract
In this manuscript, we have presented the development of a novel platform physiologically-based pharmacokinetic (PBPK) model to characterize brain disposition of mAbs in the mouse, rat, monkey and human. The model accounts for known anatomy and physiology of the brain, including the presence of distinct blood-brain barrier and blood-cerebrospinal fluid (CSF) barrier. CSF and interstitial fluid turnover, and FcRn mediated transport of mAbs are accounted for. The model was first used to characterize published and in-house pharmacokinetic (PK) data on the disposition of mAbs in rat brain, including the data on PK of mAb in different regions of brain determined using microdialysis. Majority of model parameters were fixed based on literature reported values, and only 3 parameters were estimated using rat data. The rat PBPK model was translated to mouse, monkey, and human, simply by changing the values of physiological parameters corresponding to each species. The translated PBPK models were validated by a priori predicting brain PK of mAbs in all three species, and comparing predicted exposures with observed data. The platform PBPK model was able to a priori predict all the validation PK profiles reasonably well (within threefold), without estimating any parameters. As such, the platform PBPK model presented here provides an unprecedented quantitative tool for prediction of mAb PK at the site-of-action in the brain, and preclinical-to-clinical translation of mAbs being developed against central nervous system (CNS) disorders. The proposed model can be further expanded to account for target engagement, disease pathophysiology, and novel mechanisms, to support discovery and development of novel CNS targeting mAbs.
Collapse
Affiliation(s)
- Hsueh-Yuan Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA
| | - Shengjia Wu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA
| | - Guy Meno-Tetang
- Quantitative Clinical Pharmacology/PK-PD, Modeling & Simulation, Immunology/Inflammation, UCB Pharmaceuticals, Brussels, Belgium
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
6
|
Nguyen TT, Angeli E, Darrouzain F, Nguyen QT, Desvignes C, Rigal M, Nevine O, Nicolas P, Le QV, Winterman S, Pailler MC, Zelek L, Paintaud G, Janin A, Bousquet G. A successful compartmental approach for the treatment of breast cancer brain metastases. Cancer Chemother Pharmacol 2019; 83:573-580. [PMID: 30610367 DOI: 10.1007/s00280-018-3752-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/04/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Brain metastases are challenging daily practice in oncology and remain a compartmental problem since most anti-cancer drugs do not cross the blood-brain barrier at relevant pharmacological concentrations. METHODS In a young woman with HER2-overexpressing breast cancer resistant to standard treatments, at the time of brain metastases progression, a ventricular reservoir was implanted for intrathecal drug injections and detailed pharmacokinetic studies. RESULTS A first association of intrathecal trastuzumab with intravenous cisplatin was offered to the patient. For trastuzumab, the mean cerebrospinal fluid trough concentration of 53.4 mg/L reached relevant levels, enabling the stabilization of the metastases. Adding intravenous cisplatin was not beneficial, since the cerebrospinal fluid exposure was almost undetectable under 0.08 mg/L. We then offered the patient an intrathecal combination of trastuzumab and methotrexate, because of their in vitro synergic cytotoxicity. The cerebrospinal fluid peak of methotrexate was 1037 µmol/L at 2 h, and the concentrations remained above the theoretical therapeutic concentration. After 2 months of this drug combination, we obtained an excellent response on the brain metastases. CONCLUSION Our preliminary study supports the interest of a compartmental approach through a direct administration of drugs into the cerebrospinal fluid for the treatment of breast cancer brain metastases.
Collapse
Affiliation(s)
- Thuy T Nguyen
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France.,AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France.,Medical Oncology Department A, National Cancer Hospital, Ha Noi, Viet Nam
| | - Eurydice Angeli
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - François Darrouzain
- Université François-Rabelais de Tours, CNRS, GICC UMR 7292, 37000, Tours, France.,CHRU de Tours, Pharmacologie-Toxicologie, 37000, Tours, France
| | - Quang T Nguyen
- Medical Oncology Department A, National Cancer Hospital, Ha Noi, Viet Nam
| | | | - Marthe Rigal
- AP-HP Hôpital Avicenne, Pharmacie, 93000, Bobigny, France
| | - Osman Nevine
- AP-HP Hôpital Avicenne, Pharmacie, 93000, Bobigny, France
| | - Patrick Nicolas
- AP-HP Hôpital Avicenne, Service de Biochimie, 93000, Bobigny, France
| | - Quang V Le
- Surgery Department A, National Cancer Hospital, Ha Noi, Viet Nam
| | - Sabine Winterman
- AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France
| | | | - Laurent Zelek
- AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France.,Université Paris 13, 93430, Villetaneuse, France
| | - Gilles Paintaud
- Université François-Rabelais de Tours, CNRS, GICC UMR 7292, 37000, Tours, France.,CHRU de Tours, Pharmacologie-Toxicologie, 37000, Tours, France
| | - Anne Janin
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France. .,AP-HP Hôpital Saint-Louis, Laboratoire de Pathologie, 75010, Paris, France. .,Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1165, 75010, Paris, France.
| | - Guilhem Bousquet
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France. .,AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France. .,Université Paris 13, 93430, Villetaneuse, France. .,Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1165, 75010, Paris, France.
| |
Collapse
|
7
|
Abstract
Leptomeningeal metastasis (LM) results from dissemination of cancer cells to both the leptomeninges (pia and arachnoid) and cerebrospinal fluid (CSF) compartment. Breast cancer, lung cancer, and melanoma are the most common solid tumors that cause LM. Recent approval of more active anticancer therapies has resulted in improvement in survival that is partly responsible for an increased incidence of LM. Neurologic deficits, once manifest, are mostly irreversible, and often have a significant impact on patient quality of life. LM-directed therapy is based on symptom palliation, circumscribed use of neurosurgery, limited field radiotherapy, intra-CSF and systemic therapies. Novel methods of detecting LM include detection of CSF circulating tumor cells and tumor cell-free DNA. A recent international guideline for a standardization of response assessment in LM may improve cross-trial comparisons as well as within-trial evaluation of treatment. An increasing number of retrospective studies suggest that molecular-targeted therapy, such as EGFR and ALK inhibitors in lung cancer, trastuzumab in HER2+ breast cancer, and BRAF inhibitors in melanoma, may be effective as part of the multidisciplinary management of LM. Prospective randomized trials with standardized response assessment are needed to further validate these preliminary findings.
Collapse
|
8
|
Barten DM, Cadelina GW, Weed MR. Dosing, collection, and quality control issues in cerebrospinal fluid research using animal models. HANDBOOK OF CLINICAL NEUROLOGY 2018; 146:47-64. [PMID: 29110779 DOI: 10.1016/b978-0-12-804279-3.00004-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cerebrospinal fluid (CSF) is a complex fluid filling the ventricular system and surrounding the brain and spinal cord. Although the bulk of CSF is created by the choroid plexus, a significant fraction derives from the interstitial fluid in the brain and spinal cord parenchyma. For this reason, CSF can often be used as a source of pharmacodynamic and prognostic biomarkers to reflect biochemical changes occurring within the brain. For instance, CSF biomarkers can be used to diagnose and track progression of disease as well as understand pharmacokinetic and pharmacodynamic relationships in clinical trials. To facilitate the use of these biomarkers in humans, studies in preclinical species are often valuable. This review summarizes methods for preclinical CSF collection for biomarkers from mice, rats, and nonhuman primates. In addition, dosing directly into CSF is increasingly being used to improve drug levels in the brain. Therefore, this review also summarizes the state of the art in CSF dosing in these preclinical species.
Collapse
Affiliation(s)
- Donna M Barten
- Genetically Defined Diseases, Bristol-Myers Squibb, Wallingford, CT, United States
| | - Gregory W Cadelina
- Genetically Defined Diseases, Bristol-Myers Squibb, Wallingford, CT, United States
| | - Michael R Weed
- Genetically Defined Diseases, Bristol-Myers Squibb, Wallingford, CT, United States; RxGen, Inc, New Haven, CT, United States.
| |
Collapse
|
9
|
Imaging, biodistribution, and toxicology evaluation of (212)Pb-TCMC-trastuzumab in nonhuman primates. Nucl Med Biol 2016; 43:391-6. [PMID: 27179247 DOI: 10.1016/j.nucmedbio.2016.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/01/2016] [Accepted: 04/06/2016] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The biodistribution and toxicology of a radiotherapeutic (212)Pb-trastuzumab conjugate were evaluated in nonhuman primates to meet the investigational new drug requirements prior to a phase I clinical trial in human subjects. METHODS Male cynomolgus monkeys (n=3/group) were injected intraperitoneally with the (212)Pb-trastuzumab conjugate and terminated at 8h, 10d, and 90d post-injection. Quantitative imaging studies in phantoms and monkeys were conducted using a planar gamma camera and a high purity germanium (HPGe) detector out to 48h following injection. Biodistribution analyses were conducted at 8h; all tissues and time points were evaluated for macroscopic and microscopic pathology. Blood samples were taken throughout the 90d study period for assessment of hematology parameters and serum chemistry parameters. RESULTS Quantitative gamma camera imaging and region-of-interest analyses of phantoms and monkeys indicated that 95.5±5.0% of the decay-corrected (212)Pb activity was retained in the peritoneal region up to 48h following administration of the (212)Pb-trastuzumab. Gamma-ray spectroscopy analyses confirmed that 87.6±4.5% of the decay-corrected (212)Bi activity was also retained in the peritoneal cavity during this time. Serum chemistry parameters for all groups always fell within normal ranges. Gross and histopathology evaluations showed no radiation-related toxicity in any tissue at any time. CONCLUSION In vivo imaging and biodistribution analyses showed that about 90% of both (212)Pb and decay product (212)Bi remained in the monkey peritoneal cavity. The imaging methods could also be applied to human subjects. The lack of toxicity observed in monkeys following intraperitoneal injection of the (212)Pb-trastuzumab conjugate supports its clinical assessment in humans.
Collapse
|
10
|
Watzlawik JO, Kahoud RJ, Ng S, Painter MM, Papke LM, Zoecklein L, Wootla B, Warrington AE, Carey WA, Rodriguez M. Polysialic acid as an antigen for monoclonal antibody HIgM12 to treat multiple sclerosis and other neurodegenerative disorders. J Neurochem 2015; 134:865-78. [PMID: 25866077 DOI: 10.1111/jnc.13121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/27/2015] [Accepted: 04/07/2015] [Indexed: 01/15/2023]
Abstract
CNS regeneration is a desirable goal for diseases of brain and spinal cord. Current therapeutic strategies for the treatment of multiple sclerosis (MS) aim to eliminate detrimental effects of the immune system, so far without reversing disability or affecting long-term prognosis in patients. Approachable molecular targets that stimulate CNS repair are not part of the clinical praxis or have not been identified yet. The purpose of this study was to identify the molecular target of the human monoclonal antibody HIgM12. HIgM12 reverses motor deficits in chronically demyelinated mice, a model of MS. Here, we identified polysialic acid (PSA) attached to the neural cell adhesion molecule (NCAM) as the antigen for HIgM12 by using different NCAM knockout strains and through PSA removal from the NCAM protein core. Antibody binding to CNS tissue and primary cells, antibody-mediated cell adhesion, and neurite outgrowth on HIgM12-coated nitrocellulose was detected only in the presence of PSA as assessed by western blotting, immunoprecipitation, immunocytochemistry, and histochemistry. We conclude that HIgM12 mediates its in vivo and in vitro effects through binding to PSA and has the potential to be an effective therapy for MS and neurodegenerative diseases. The human antibody HIgM12 stimulates neurite outgrowth in vitro and promotes function in chronically demyelinated mice, a model of multiple sclerosis. The cellular antigen for HIgM12 was undetermined. Here, we identified polysialic acid attached to NCAM (neural cell adhesion molecule) as the cellular target for HIgM12. This includes glial fibrillary acidic protein (GFAP)-positive mouse astrocytes (GFAP, red; HIgM12, green; DAPI, blue) among other cell types of the central nervous system. These findings indicate a new strategy for the treatment of neuro-motor disorders including multiple sclerosis.
Collapse
Affiliation(s)
- Jens O Watzlawik
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Robert J Kahoud
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.,Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Shermayne Ng
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Meghan M Painter
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Louisa M Papke
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Laurie Zoecklein
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Bharath Wootla
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - William A Carey
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Stanimirovic D, Kemmerich K, Haqqani AS, Farrington GK. Engineering and pharmacology of blood-brain barrier-permeable bispecific antibodies. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:301-35. [PMID: 25307221 DOI: 10.1016/bs.apha.2014.06.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The development and approval of antibody-based therapeutics have progressed rapidly over the past decade. However, poor blood-brain barrier (BBB) permeability hinders the progress of antibody therapies for conditions in which the target is located in the central nervous system (CNS). Increased brain penetration of therapeutic antibodies can be achieved by engineering bispecific antibodies in which one antibody binding specificity recognizes a BBB receptor that undergoes receptor-mediated transcytosis (RMT) from the circulatory compartment into brain parenchyma, and the second binding specificity recognizes a therapeutic target within the CNS. These bispecific antibodies can be built using various antibody fragments as "building blocks," including monomeric single-domain antibodies, the smallest antigen-binding fragments of immunoglobulins. The development of BBB-crossing bispecific antibodies requires targeted antibody engineering to optimize multiple characteristics of "BBB carrier" and therapeutic arms, as well as other antibody properties impacting pharmacokinetics and effector function. Whereas several BBB-crossing bispecific antibodies have been developed using transferrin receptor antibodies as BBB carriers, the principal obstacle for capitalizing on the future promise of CNS-active antibodies remains the scarcity of known, characterized RMT receptors which could be exploited for the development of BBB carriers. This chapter reviews the recent advances and guiding principles for designing, engineering, and evaluating BBB-crossing bispecific antibodies and discusses approaches to identify and characterize novel BBB-crossing antibodies and RMT receptors.
Collapse
Affiliation(s)
- Danica Stanimirovic
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada.
| | - Kristin Kemmerich
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Arsalan S Haqqani
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | | |
Collapse
|
12
|
Calias P, Banks WA, Begley D, Scarpa M, Dickson P. Intrathecal delivery of protein therapeutics to the brain: a critical reassessment. Pharmacol Ther 2014; 144:114-22. [PMID: 24854599 DOI: 10.1016/j.pharmthera.2014.05.009] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/12/2014] [Indexed: 12/25/2022]
Abstract
Disorders of the central nervous system (CNS), including stroke, neurodegenerative diseases, and brain tumors, are the world's leading causes of disability. Delivery of drugs to the CNS is complicated by the blood-brain barriers that protect the brain from the unregulated leakage and entry of substances, including proteins, from the blood. Yet proteins represent one of the most promising classes of therapeutics for the treatment of CNS diseases. Many strategies for overcoming these obstacles are in development, but the relatively straightforward approach of bypassing these barriers through direct intrathecal administration has been largely overlooked. Originally discounted because of its lack of usefulness for delivering small, lipid-soluble drugs to the brain, the intrathecal route has emerged as a useful, in some cases perhaps the ideal, route of administration for certain therapeutic protein and targeted disease combinations. Here, we review blood-brain barrier functions and cerebrospinal fluid dynamics and their relevance to drug delivery via the intrathecal route, discuss animal and human studies that have investigated intrathecal delivery of protein therapeutics, and outline several characteristics of protein therapeutics that can allow them to be successfully delivered intrathecally.
Collapse
Affiliation(s)
| | - William A Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care Center, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - David Begley
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Maurizio Scarpa
- Department of Paediatrics, University of Padova, Padova, Italy
| | - Patricia Dickson
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
13
|
Freyer CW, Yaghmour G, Jennings K, Dhanapal V. Drug-Induced Aseptic Meningitis Associated With Intrathecal Trastuzumab. J Pharm Technol 2014; 30:43-47. [PMID: 34860873 PMCID: PMC5990126 DOI: 10.1177/8755122513500918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2024] Open
Abstract
Objective: To report a case of drug-induced aseptic meningitis (DIAM) in a patient receiving intrathecal trastuzumab (ITT) for leptomeningeal carcinomatosis (LC) secondary to HER-2/neu positive breast cancer. Case Summary: A 43-year-old female with stage IV breast cancer presented with headache, parasthesias, and aphasia. Brain imaging suggested leptomeningeal enhancement. Adenocarcinoma cells were found on cerebrospinal fluid analysis, and infectious etiologies were excluded. The patient received 30 mg of ITT via Ommaya reservoir and suffered sudden neurologic deterioration within 2 hours. Given the sudden onset of clinical deterioration after ITT administration, it was determined that the patient had suffered from DIAM. The patient suffered progressive neurologic decline and was unable to care for herself any further. Discussion: Treatment for LC remains challenging due to limited clinical experience and the challenging location of the disease. ITT has been used in multiple reports without adverse events. A temporal relationship existed between ITT administration and significant neurologic deterioration, possibly related to DIAM. Our patient was not exposed to any more common causative agents of DIAM. Reasons for her lack of recovery are likely multifactorial. Symptoms of DIAM may have in part been exacerbated by progressive LC; however, prior to ITT, symptoms had shown slow but persistent progression rather than significant, acute changes in severity. Retrial of the offending agent was not attempted due to patient risk; thus, a cause-and-effect relationship cannot be established. This event is a "possible" drug-induced adverse event scoring 2 on the Naranjo algorithm. Conclusions: Use of ITT for LC has been frequently reported in recent literature with substantial efficacy and lack of adverse events. This is the first published report of any significant adverse event associated with ITT. Clinicians need to be aware of the possibility of DIAM with ITT and explore options to prevent or manage this severe complication.
Collapse
|
14
|
Le Rhun E, Taillibert S, Chamberlain MC. Carcinomatous meningitis: Leptomeningeal metastases in solid tumors. Surg Neurol Int 2013; 4:S265-88. [PMID: 23717798 PMCID: PMC3656567 DOI: 10.4103/2152-7806.111304] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/11/2013] [Indexed: 11/04/2022] Open
Abstract
Leptomeningeal metastasis (LM) results from metastatic spread of cancer to the leptomeninges, giving rise to central nervous system dysfunction. Breast cancer, lung cancer, and melanoma are the most frequent causes of LM among solid tumors in adults. An early diagnosis of LM, before fixed neurologic deficits are manifest, permits earlier and potentially more effective treatment, thus leading to a better quality of life in patients so affected. Apart from a clinical suspicion of LM, diagnosis is dependent upon demonstration of cancer in cerebrospinal fluid (CSF) or radiographic manifestations as revealed by neuraxis imaging. Potentially of use, though not commonly employed, today are use of biomarkers and protein profiling in the CSF. Symptomatic treatment is directed at pain including headache, nausea, and vomiting, whereas more specific LM-directed therapies include intra-CSF chemotherapy, systemic chemotherapy, and site-specific radiotherapy. A special emphasis in the review discusses novel agents including targeted therapies, that may be promising in the future management of LM. These new therapies include anti-epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors erlotinib and gefitinib in nonsmall cell lung cancer, anti-HER2 monoclonal antibody trastuzumab in breast cancer, anti-CTLA4 ipilimumab and anti-BRAF tyrosine kinase inhibitors such as vermurafenib in melanoma, and the antivascular endothelial growth factor monoclonal antibody bevacizumab are currently under investigation in patients with LM. Challenges of managing patients with LM are manifold and include determining the appropriate patients for treatment as well as the optimal route of administration of intra-CSF drug therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Breast Unit, Department of Medical Oncology, Centre Oscar Lambret and Department of Neuro Oncology, Roger Salengro Hospital, University Hospital, Lille, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Research Cancer Center, Seattle, WA, USA
| |
Collapse
|
15
|
Caffo M, Barresi V, Caruso G, Cutugno M, La Fata G, Venza M, Alafaci C, Tomasello F. Innovative therapeutic strategies in the treatment of brain metastases. Int J Mol Sci 2013; 14:2135-74. [PMID: 23340652 PMCID: PMC3565370 DOI: 10.3390/ijms14012135] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 01/08/2013] [Accepted: 01/09/2013] [Indexed: 12/29/2022] Open
Abstract
Brain metastases (BM) are the most common intracranial tumors and their incidence is increasing. Untreated brain metastases are associated with a poor prognosis and a poor performance status. Metastasis development involves the migration of a cancer cell from the bulk tumor into the surrounding tissue, extravasation from the blood into tissue elsewhere in the body, and formation of a secondary tumor. In the recent past, important results have been obtained in the management of patients affected by BM, using surgery, radiation therapy, or both. Conventional chemotherapies have generally produced disappointing results, possibly due to their limited ability to penetrate the blood-brain barrier. The advent of new technologies has led to the discovery of novel molecules and pathways that have better depicted the metastatic process. Targeted therapies such as bevacizumab, erlotinib, gefitinib, sunitinib and sorafenib, are all licensed and have demonstrated improved survival in patients with metastatic disease. In this review, we will report current data on targeted therapies. A brief review about brain metastatic process will be also presented.
Collapse
Affiliation(s)
- Maria Caffo
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
| | - Valeria Barresi
- Department of Human Pathology, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mail:
| | - Gerardo Caruso
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +39-090-2217167; Fax: +39-090-693714
| | - Mariano Cutugno
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
| | - Giuseppe La Fata
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
| | - Mario Venza
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
| | - Concetta Alafaci
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
| | - Francesco Tomasello
- Department of Neurosciences, School of Medicine, University of Messina, A.O.U. Policlinico “G. Martino”, via Consolare Valeria, 1, 98125 Messina, Italy; E-Mails: (M.C.); (M.C.); (G.L.F.); (M.V.); (C.A.); (F.T.)
| |
Collapse
|
16
|
Vargas HM, Amouzadeh HR, Engwall MJ. Nonclinical strategy considerations for safety pharmacology: evaluation of biopharmaceuticals. Expert Opin Drug Saf 2012; 12:91-102. [DOI: 10.1517/14740338.2013.745851] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
17
|
Daniels TR, Martínez-Maza O, Penichet ML. Animal models for IgE-meditated cancer immunotherapy. Cancer Immunol Immunother 2011; 61:1535-46. [PMID: 22193986 DOI: 10.1007/s00262-011-1169-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 11/17/2011] [Indexed: 10/14/2022]
Abstract
Although most monoclonal antibodies developed for cancer therapy are of the IgG class, antibodies of the IgE class have certain properties that make them attractive as cancer therapeutics. These properties include the superior affinity for the Fc epsilon receptors (FcεRs), the low serum level of IgE that minimizes competition of endogenous IgE for FcεR occupancy, and the ability to induce a broad and vigorous immune response through the interaction with multiple cells including mast cells, basophils, monocytes, macrophages, dendritic cells, and eosinophils. Tumor-targeted IgE antibodies are expected to harness the allergic response against tumors and activate a secondary, T-cell-mediated immune response. Importantly, the IgE antibody can be used for passive immunotherapy and as an adjuvant of cancer vaccines. However, there are important limitations in the use of animal models including the fact that human IgE does not interact with rodent FcεRs and that there is a different cellular distribution of FcεRs in humans and rodents. Despite these limitations, different murine models have been used with success to evaluate the in vivo anti-cancer activity of several IgE antibodies. These models include wild-type immunocompetent animals bearing syngeneic tumors, xenograft models using immunocompromised mice bearing human tumors and reconstituted with human effector cells, and human FcεRIα transgenic mice bearing syngeneic tumors. In addition, non-human primates such as cynomolgus monkeys can be potentially used for toxicological and pharmacokinetic studies. This article describes the advantages and disadvantages of these models and their use in evaluating the in vivo properties of IgE antibodies for cancer therapy.
Collapse
Affiliation(s)
- Tracy R Daniels
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, CHS 54-140, Box 951782, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
18
|
Wang Y, Han ZB, Ma J, Zuo C, Geng J, Gong W, Sun Y, Li H, Wang B, Zhang L, He Y, Han ZC. A toxicity study of multiple-administration human umbilical cord mesenchymal stem cells in cynomolgus monkeys. Stem Cells Dev 2011; 21:1401-8. [PMID: 21958114 DOI: 10.1089/scd.2011.0441] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Therapies based on stem cells have shown very attractive potential in many clinical studies. However, the data about the safety of stem cells application remains insufficient. The present study was designed to evaluate the overall toxicology of human umbilical cord mesenchymal stem cells (hUC-MSCs) in cynomolgus monkeys with repeated administrations. hUC-MSCs were administered by intravenous injection once every 2 weeks for 6 weeks. The dose levels employed in this study were 2×10(6), 1×10(7) cells/kg body weight. Toxicity was evaluated by clinical observations (body weight, body temperature, and ophthalmology exams), pathology (blood cell counts, clinical biochemistry, urinalysis, and bone marrow smears), immunologic consequences (lymphoproliferation assay, the secretion of interferon-γ and interleukin-4, the percentage of CD3, CD4, CD8 T cells, and the ratio of CD4 and CD8 T cells) and anatomic pathology. Pharmacodynamics in blood and distribution of hUC-MSCs in the tissues of cynomolgus monkeys were measured by real-time polymerase chain reaction. All animals survived until scheduled euthanasia. No stem cells transplantation-related toxicity was found in this study. hUC-MSCs could be found in the blood of cynomolgus monkeys beyond 8 h. The findings of this 6-week toxicity study showed that the transplantation of hUC-MSCs did not affect the general health of cynomolgus monkeys.
Collapse
Affiliation(s)
- Youwei Wang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Hematology and Blood Diseases Hospital, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pepinsky RB, Shao Z, Ji B, Wang Q, Meng G, Walus L, Lee X, Hu Y, Graff C, Garber E, Meier W, Mi S. Exposure Levels of Anti-LINGO-1 Li81 Antibody in the Central Nervous System and Dose-Efficacy Relationships in Rat Spinal Cord Remyelination Models after Systemic Administration. J Pharmacol Exp Ther 2011; 339:519-29. [DOI: 10.1124/jpet.111.183483] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
20
|
Papadatos-Pastos D, Banerji U. Revisiting the role of molecular targeted therapies in patients with brain metastases. J Neurooncol 2011; 105:467-74. [DOI: 10.1007/s11060-011-0661-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 07/02/2011] [Indexed: 12/20/2022]
|
21
|
Perissinotti AJ, Reeves DJ. Role of Intrathecal Rituximab and Trastuzumab in the Management of Leptomeningeal Carcinomatosis. Ann Pharmacother 2010; 44:1633-40. [DOI: 10.1345/aph.1p197] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Objective: To review evidence for the use of intrathecal rituximab and trastuzumab in the management of leptomeningeal carcinomatosis. Data Sources: A search of MEDLINE (1966–July 2010) and International Pharmaceutical Abstracts (1970–July 2010) was performed using search terms intrathecal, trastuzumab, rituximab, and monoclonal antibody. Additionally, American Society of Clinical Oncology, San Antonio Breast Conference, American Association for Cancer Research, and American Society of Hematology meeting abstracts were searched. Study Selection and Data Extraction: Publications were reviewed for inclusion. Those reporting use of rituximab and trastuzumab intrathecally are reviewed and include 1 Phase 1 trial, 2 small prospective studies, 1 case series, and 15 case reports. Data Synthesis: The treatment of leptomeningeal carcinomatosis is challenging due to the presence of the blood–brain barrier. Numerous systemically administered therapies do not readily penetrate into the site of leptomeningeal disease and have been ineffective. Intrathecal administration of 2 monoclonal antibodies (trastuzumab and rituximab) has been investigated in case reports and case series. Additionally, intrathecal rituximab has been investigated in a Phase 1 study. Survival after intrathecal trastuzumab ranged from 39 days to greater than 72 months and the drug was well tolerated, with no adverse events attributed to it. Doses used in these reports ranged from 5 to 100 mg. Survival after intrathecal rituximab ranged from 1.1 weeks to greater than 3.5 years. In the Phase 1 trial, the maximum tolerated rituximab dose was 25 mg and 60% of patients responded. Four of the 6 responding patients experienced a complete response. Intrathecal rituximab exhibited minor toxicities that resolved quickly without long-term effects. Conclusions: Reports suggest that both trastuzumab and rituximab may be utilized intrathecally. Patients with refractory leptomeningeal carcinomatosis may benefit from a trial of intrathecal trastuzumab or rituximab; however, their use remains investigational, as more data and experience are necessary before intrathecal administration can be considered standard.
Collapse
Affiliation(s)
- Anthony J Perissinotti
- Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Ml; now, PGY1 Pharmacy Practice Resident, Department of Pharmacy, Harper University Hospital, Detroit
| | - David J Reeves
- Department of Pharmacy, Karmanos Cancer Center, Detroit; now, Oncology Clinical Pharmacist, Department of Pharmacy Services, St. Vincent Hospital, Indianapolis, IN, and Assistant Professor, Pharmacy Practice, College of Pharmacy and Health Sciences, Butler University, Indianapolis
| |
Collapse
|