1
|
von Roemeling C, Ferreri AJM, Soussain C, Tun HW, Grommes C. Targets and treatments in primary CNS lymphoma. Leuk Lymphoma 2024; 65:1055-1067. [PMID: 38659230 DOI: 10.1080/10428194.2024.2342560] [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: 02/29/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and highly aggressive lymphoma entirely localized in the central nervous system or vitreoretinal space. PCNSL generally initially responds to methotrexate-containing chemotherapy regimens, but progressive or relapsing disease is common, and the prognosis is poor for relapsed or refractory (R/R) patients. PCNSL is often characterized by activation of nuclear factor kappa B (NF-κB) due to mutations in the B-cell receptor (BCR) or toll-like receptor (TLR) pathways, as well as immune evasion. Targeted treatments that inhibit key PCNSL mechanisms and pathways are being evaluated; inhibition of Bruton's tyrosine kinase (BTK) downstream of BCR activation has demonstrated promising results in treating R/R disease. This review will summarize the evidence and potential for targeted therapeutic agents to improve treatment outcomes in PCNSL. This includes immunotherapeutic and immunomodulatory approaches and inhibitors of the key pathways driving PCNSL, such as aberrant BCR and TLR signaling.
Collapse
Affiliation(s)
- Christina von Roemeling
- Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Andrés J M Ferreri
- Department of Onco-Hematology, University Vita-Salute San Raffaele, Milano, Italy
- Department of Onco-Hematology, Lymphoma Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Carole Soussain
- Institut Curie, Service d'Hématologie, site de Saint-Cloud, France
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Han W Tun
- Department of Hematology, Mayo Clinic, Jacksonville, Florida, USA
| | - Christian Grommes
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
2
|
Reda M, Jabbour R, Haydar A, Jaafar F, El Ayoubi N, Nawfal O, Beydoun A. Case report: Rapid recovery after intrathecal rituximab administration in refractory anti-NMDA receptor encephalitis: report of two cases. Front Immunol 2024; 15:1369587. [PMID: 38510253 PMCID: PMC10950913 DOI: 10.3389/fimmu.2024.1369587] [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: 01/12/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Background Anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis is one of the most prevalent etiologies of autoimmune encephalitis. Approximately 25% of anti-NMDAR encephalitis cases prove refractory to both first- and second-line treatments, posing a therapeutic dilemma due to the scarcity of evidence-based data for informed decision-making. Intravenous rituximab is commonly administered as a second-line agent; however, the efficacy of its intrathecal administration has rarely been reported. Case summary We report two cases of severe anti-NMDAR encephalitis refractory to conventional therapies. These patients presented with acute-onset psychosis progressing to a fulminant picture of encephalitis manifesting with seizures, dyskinesia, and dysautonomia refractory to early initiation of first- and second-line therapeutic agents. Both patients received 25 mg of rituximab administered intrathecally, repeated weekly for a total of four doses, with no reported adverse effects. Improvement began 2-3 days after the first intrathecal administration, leading to a dramatic recovery in clinical status and functional performance. At the last follow-up of 6 months, both patients remain in remission without the need for maintenance immunosuppression. Conclusion Our cases provide evidence supporting the intrathecal administration of rituximab as a therapeutic option for patients with refractory anti-NMDAR encephalitis. Considering the limited penetration of intravenous rituximab into the central nervous system, a plausible argument can be made favoring intrathecal administration as the preferred route or the simultaneous administration of intravenous and intrathecal rituximab. This proposition warrants thorough investigation in subsequent clinical trials.
Collapse
Affiliation(s)
- Mahasen Reda
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rosette Jabbour
- Division of Neurology, Saint George Hospital University Medical Center, Beirut, Lebanon
| | - Asad Haydar
- Division of Neurology, Saint George Hospital University Medical Center, Beirut, Lebanon
| | - Fatima Jaafar
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nabil El Ayoubi
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Omar Nawfal
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ahmad Beydoun
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
3
|
Schellhammer L, Beffinger M, Salazar U, Laman JD, Buch T, vom Berg J. Exit pathways of therapeutic antibodies from the brain and retention strategies. iScience 2023; 26:108132. [PMID: 37915602 PMCID: PMC10616392 DOI: 10.1016/j.isci.2023.108132] [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] [Indexed: 11/03/2023] Open
Abstract
Treating brain diseases requires therapeutics to pass the blood-brain barrier (BBB) which is nearly impermeable for large biologics such as antibodies. Several methods now facilitate crossing or circumventing the BBB for antibody therapeutics. Some of these exploit receptor-mediated transcytosis, others use direct delivery bypassing the BBB. However, successful delivery into the brain does not preclude exit back to the systemic circulation. Various mechanisms are implicated in the active and passive export of antibodies from the central nervous system. Here we review findings on active export via transcytosis of therapeutic antibodies - in particular, the role of the neonatal Fc receptor (FcRn) - and discuss a possible contribution of passive efflux pathways such as lymphatic and perivascular drainage. We point out open questions and how to address these experimentally. In addition, we suggest how emerging findings could aid the design of the next generation of therapeutic antibodies for neurologic diseases.
Collapse
Affiliation(s)
- Linda Schellhammer
- Institute of Laboratory Animal Science, University of Zurich, 8952 Schlieren, Switzerland
| | - Michal Beffinger
- Institute of Laboratory Animal Science, University of Zurich, 8952 Schlieren, Switzerland
- InCephalo AG, 4123 Allschwil, Switzerland
| | - Ulisse Salazar
- Institute of Laboratory Animal Science, University of Zurich, 8952 Schlieren, Switzerland
| | - Jon D. Laman
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9713, the Netherlands
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, 8952 Schlieren, Switzerland
| | - Johannes vom Berg
- Institute of Laboratory Animal Science, University of Zurich, 8952 Schlieren, Switzerland
- InCephalo AG, 4123 Allschwil, Switzerland
| |
Collapse
|
4
|
Zhang Y, Liu Z, Gao C, Bian H, Ma Y, Jing F, Zhao X. Role of Rituximab in Treatment of Patients With Primary Central Nervous System Lymphoma (PCNSL): A Systematic Review and Meta-Analysis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:733-741. [PMID: 37453867 DOI: 10.1016/j.clml.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/10/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare form of non-Hodgkin's lymphoma involving the brain, cerebrospinal fluid, spinal cord and eyes. Rituximab has played a prominent role in the treatment of non-Hodgkin's B-cell lymphomas, including aggressive diffuse large B lymphoma. However, as a macromolecular drug, the role of rituximab in the treatment of PCNSL has been controversial. In this systematic review and meta-analysis, we evaluated the role of rituximab in the treatment of PCNSL. We searched articles in the following electronic databases including PubMed, Embase, Cochrane Library, Web of Science, and ClinicalTrials.gov until October 20, 2022.We included 11 studies (3 RCTS and 8 retrospective studies) with a total of 1182 patients. We extracted the baseline characteristics and outcomes of the studies and assessed the risk of bias, then used Review Manager 5.4 for this meta-analysis. The primary outcomes included complete response rate (CR), overall survival (OS), and progression-free survival (PFS). Odds ratios (ORS) and corresponding 95% confidence intervals (CIS) for the primary outcome were analyzed and compared. The results of our statistical analysis show that the use of rituximab was closely correlated with a higher CR(OR 1.70,95%CI 1.17-2.46, P = .005), 3-year OS (OR 2.40, 95%CI 1.53-3.77, P = .0001), 5-year OS (OR 2.75, 95%CI 1.68-4.49, P < .0001), 3-year PFS(OR 4.42, 95%CI 1.15-16.97, P < .0001), 5-year PFS(OR 1.97, 95%CI 1.39-2.78, P = .0001).These results suggest that rituximab may have a positive impact on the prognosis of patients with PCNSL, and may be helpful in the determination of treatment plan for patients with PCNSL.
Collapse
Affiliation(s)
- Yuhang Zhang
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Zhihe Liu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Chengwen Gao
- Laboratory of Medical Biology, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Haiyan Bian
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yushuo Ma
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Fanjing Jing
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Xia Zhao
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China.
| |
Collapse
|
5
|
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
|
6
|
Che J, DePalma TJ, Sivakumar H, Mezache LS, Tallman MM, Venere M, Swindle-Reilly K, Veeraraghavan R, Skardal A. αCT1 peptide sensitizes glioma cells to temozolomide in a glioblastoma organoid platform. Biotechnol Bioeng 2023; 120:1108-1119. [PMID: 36544242 DOI: 10.1002/bit.28313] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Glioblastoma (GBM) is the most common form of brain cancer. Even with aggressive treatment, tumor recurrence is almost universal and patient prognosis is poor because many GBM cell subpopulations, especially the mesenchymal and glioma stem cell populations, are resistant to temozolomide (TMZ), the most commonly used chemotherapeutic in GBM. For this reason, there is an urgent need for the development of new therapies that can more effectively treat GBM. Several recent studies have indicated that high expression of connexin 43 (Cx43) in GBM is associated with poor patient outcomes. It has been hypothesized that inhibition of the Cx43 hemichannels could prevent TMZ efflux and sensitize otherwise resistance cells to the treatment. In this study, we use a three-dimensional organoid model of GBM to demonstrate that combinatorial treatment with TMZ and αCT1, a Cx43 mimetic peptide, significantly improves treatment efficacy in certain populations of GBM. Confocal imaging was used to visualize changes in Cx43 expression in response to combinatorial treatment. These results indicate that Cx43 inhibition should be pursued further as an improved treatment for GBM.
Collapse
Affiliation(s)
- Jingru Che
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Thomas J DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | - Louisa S Mezache
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Miranda M Tallman
- Dorothy M. Davis Hearth and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Radiation Oncology, James Cancer Hospital and Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Monica Venere
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Radiation Oncology, James Cancer Hospital and Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Katelyn Swindle-Reilly
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, Ohio, USA
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
- The Ohio State University and Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Center for Cancer Engineering, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
7
|
Vitanza NA, Ronsley R, Choe M, Henson C, Breedt M, Barrios-Anderson A, Wein A, Brown C, Beebe A, Kong A, Kirkey D, Lee BM, Leary SES, Crotty EE, Hoeppner C, Holtzclaw S, Wilson AL, Gustafson JA, Foster JB, Iliff JJ, Goldstein HE, Browd SR, Lee A, Ojemann JG, Pinto N, Gust J, Gardner RA, Jensen MC, Hauptman JS, Park JR. Locoregional CAR T cells for children with CNS tumors: Clinical procedure and catheter safety. Neoplasia 2023; 36:100870. [PMID: 36599192 PMCID: PMC9823206 DOI: 10.1016/j.neo.2022.100870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023]
Abstract
Central nervous system (CNS) tumors are the most common solid malignancy in the pediatric population. Based on adoptive cellular therapy's clinical success against childhood leukemia and the preclinical efficacy against pediatric CNS tumors, chimeric antigen receptor (CAR) T cells offer hope of improving outcomes for recurrent tumors and universally fatal diseases such as diffuse intrinsic pontine glioma (DIPG). However, a major obstacle for tumors of the brain and spine is ineffective T cell chemotaxis to disease sites. Locoregional CAR T cell delivery via infusion through an intracranial catheter is currently under study in multiple early phase clinical trials. Here, we describe the Seattle Children's single-institution experience including the multidisciplinary process for the preparation of successful, repetitive intracranial T cell infusion for children and the catheter-related safety of our 307 intracranial CAR T cell doses.
Collapse
Affiliation(s)
- Nicholas A Vitanza
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Rebecca Ronsley
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michelle Choe
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Casey Henson
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Mandy Breedt
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Adriel Barrios-Anderson
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Amy Wein
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Christopher Brown
- Seattle Children's Therapeutics, Seattle, WA, USA; Therapeutic Cell Production Core, Seattle Children's Research Institute, Seattle, WA, USA
| | - Adam Beebe
- Seattle Children's Therapeutics, Seattle, WA, USA; Therapeutic Cell Production Core, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ada Kong
- Department of Pharmacy, Seattle Children's Hospital, Seattle, WA, USA
| | - Danielle Kirkey
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Brittany M Lee
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Sarah E S Leary
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Erin E Crotty
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Corrine Hoeppner
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Susan Holtzclaw
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | | | | | - Jessica B Foster
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA; Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA; Division of Pediatric Neurology, Department of Neurology, University of Washington, Seattle, WA, USA
| | - Hannah E Goldstein
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Samuel R Browd
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Amy Lee
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Jeffrey G Ojemann
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Navin Pinto
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Juliane Gust
- Division of Pediatric Neurology, Department of Neurology, University of Washington, Seattle, WA, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Rebecca A Gardner
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Seattle Children's Therapeutics, Seattle, WA, USA
| | | | - Jason S Hauptman
- Division of Neurosurgery, Seattle Children's Hospital & Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Julie R Park
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Seattle Children's Therapeutics, Seattle, WA, USA
| |
Collapse
|
8
|
Li S, Wang C, Chen J, Lan Y, Zhang W, Kang Z, Zheng Y, Zhang R, Yu J, Li W. Signaling pathways in brain tumors and therapeutic interventions. Signal Transduct Target Ther 2023; 8:8. [PMID: 36596785 PMCID: PMC9810702 DOI: 10.1038/s41392-022-01260-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 01/05/2023] Open
Abstract
Brain tumors, although rare, contribute to distinct mortality and morbidity at all ages. Although there are few therapeutic options for brain tumors, enhanced biological understanding and unexampled innovations in targeted therapies and immunotherapies have considerably improved patients' prognoses. Nonetheless, the reduced response rates and unavoidable drug resistance of currently available treatment approaches have become a barrier to further improvement in brain tumor (glioma, meningioma, CNS germ cell tumors, and CNS lymphoma) treatment. Previous literature data revealed that several different signaling pathways are dysregulated in brain tumor. Importantly, a better understanding of targeting signaling pathways that influences malignant behavior of brain tumor cells might open the way for the development of novel targeted therapies. Thus, there is an urgent need for a more comprehensive understanding of the pathogenesis of these brain tumors, which might result in greater progress in therapeutic approaches. This paper began with a brief description of the epidemiology, incidence, risk factors, as well as survival of brain tumors. Next, the major signaling pathways underlying these brain tumors' pathogenesis and current progress in therapies, including clinical trials, targeted therapies, immunotherapies, and system therapies, have been systemically reviewed and discussed. Finally, future perspective and challenges of development of novel therapeutic strategies in brain tumor were emphasized.
Collapse
Affiliation(s)
- Shenglan Li
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Can Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinyi Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanjie Lan
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weichunbai Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuang Kang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yi Zheng
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rong Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianyu Yu
- grid.24696.3f0000 0004 0369 153XDepartment of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Treatment Options for Recurrent Primary CNS Lymphoma. Curr Treat Options Oncol 2022; 23:1548-1565. [PMID: 36205806 DOI: 10.1007/s11864-022-01016-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT Primary CNS lymphoma (PCNSL) constitutes a rare extranodal variant of non-Hodgkin lymphoma (NHL) with an annual incidence of 0.45/100,000. Given the paucity of large prospective clinical trials, there is no consensus treatment for refractory or relapsed (r/r) PCNSL, and available strategies are largely based on retrospective analyses. Patient age, performance status, previously administered treatment, duration of response, and molecular characteristics guide selection of salvage therapy. Patients with a good performance status (KPS >70), particularly ≤65 years, and adequate organ function should be considered for salvage polychemotherapy. Based on its high overall response rate even in the relapsed setting, we choose high-dose (≥ 3.5g/m2) methotrexate (HD-MTX) based regimens, e.g., R-MPV (rituximab, HD-MTX, procarbazine, and vincristine), for remission re-induction as long as patients were sensitive to first line HD-MTX-based regimens, especially when duration of previous response was ≥ 1 year. Following successful remission induction, we choose myeloablative chemotherapy (e.g., thiotepa, busulfan, cyclophosphamide) and subsequent autologous stem cell transplant in curative intent whenever feasible. Alternatively, conventional chemotherapy regimens (for example, monthly HD-MTX) or low-dose whole-brain radiation therapy (WBRT) are selected for consolidation in non-transplant candidates in complete remission. In cases of HD-MTX refractory disease or contraindications, we use pemetrexed; temozolomide/rituximab; high-dose cytarabine; or whole brain radiation for remission induction. Clinical trial participation is considered as well. Emerging therapies for upfront or salvage therapy under ongoing investigation include bruton tyrosine kinase inhibition (e.g., ibrutinib), immunomodulatory drugs (e.g., lenalidomide), immune checkpoint inhibitors (ICI, e.g., nivolumab), and chimeric antigen receptor T (CAR-T) cell therapy.
Collapse
|
10
|
Secondary Central Nervous System Lymphoma: Updates in Treatment and Prophylaxis Strategies. Curr Treat Options Oncol 2022; 23:1443-1456. [PMID: 36127571 DOI: 10.1007/s11864-022-01017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2022] [Indexed: 11/03/2022]
Abstract
OPINION STATEMENT Referring to any central nervous system (CNS) involvement with preceding or concurrent systemic disease, secondary CNS lymphoma (SCNSL) lacks a clear standard of care and historically carries a very poor prognosis. Aggressive histologies predominate, most notably diffuse large B cell lymphoma (DLBCL), with higher relative frequency in Burkitt lymphoma but lower absolute incidence. Therapeutic strategies commonly feature intensive CNS-penetrant chemotherapy, including methotrexate, cytarabine, and others. Combination regimens, novel targeted agents, and cellular therapy considerations are reviewed, noting that patients with SCNSL are often excluded from clinical trials and dedicated SCNSL studies are historically limited. Given these challenges, there has been renewed attention on CNS prophylaxis as well as strategies for early CNS detection. Prophylaxis is standard of care in Burkitt lymphoma, whereas its role in DLBCL and related histologies is increasingly unclear.
Collapse
|
11
|
Khorasanchi A, Benson Z, Hall M, Ebadirad N, Gharavi MH, Willard P, Chimzar M, McKay J, Simmons G, Yazbeck V. Systemic Relapse in a Young Adult Patient with Primary CNS Diffuse Large B-Cell Lymphoma. Case Rep Hematol 2022; 2022:7139661. [PMID: 35774056 PMCID: PMC9239761 DOI: 10.1155/2022/7139661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/06/2022] [Indexed: 12/11/2022] Open
Abstract
Primary central nervous system diffuse large B-cell lymphoma (PCNS-DLBCL) is a rare form of non-Hodgkin's lymphoma, characterized by an aggressive disease course. While CNS relapse is common, systemic relapse is rare with no consensus on optimal treatment. This paper presents an unusual case of advanced PCNS-DLBCL with systemic relapse, including adrenal gland involvement. A review of the existing literature and a discussion on the management of systemic relapse in PCNS-DLBCL is also provided.
Collapse
Affiliation(s)
- Adam Khorasanchi
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Benson
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Misty Hall
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Nelya Ebadirad
- Department of Radiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohammad H. Gharavi
- Department of Radiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Patrick Willard
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Miranda Chimzar
- School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - John McKay
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Gary Simmons
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Victor Yazbeck
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
12
|
Kaulen LD, Gumbinger C, Hinz F, Kessler T, Winkler F, Bendszus M, Sahm F, Wick W. Intraventricular immune checkpoint inhibition with nivolumab in relapsed primary central nervous system lymphoma. Neurooncol Adv 2022; 4:vdac051. [PMID: 35571985 PMCID: PMC9092640 DOI: 10.1093/noajnl/vdac051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
SUMMARY
Intrathecal nivolumab combined with its systemic administration was recently found safe and effective in melanoma with leptomeningeal dissemination, prompting us to evaluate intraventricular nivolumab for recurrent primary CNS lymphoma (PCNSL) in an elderly patient unable to tolerate aggressive systemic polychemotherapy. Intraventricular nivolumab achieved a lasting (>12 months) complete remission including parenchymal lesions distant from cerebrospinal fluid spaces. No toxicities or adverse events related to the mode of administration were noted. Our case suggests intraventricular nivolumab is active in recurrent parenchymal PCNSL. Together with detected 9p24.1 gains this argues for further prospective evaluation, for which our treatment protocol provides a framework.
Collapse
Affiliation(s)
- Leon D Kaulen
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Gumbinger
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Hinz
- Department of Neuropathology, Heidelberg University Hospital, and Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tobias Kessler
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Winkler
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, and Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
13
|
Sadekar SS, Bowen M, Cai H, Jamalian S, Rafidi H, Shatz‐Binder W, Lafrance‐Vanasse J, Chan P, Meilandt WJ, Oldendorp A, Sreedhara A, Daugherty A, Crowell S, Wildsmith KR, Atwal J, Fuji RN, Horvath J. Translational approaches for brain delivery of biologics via cerebrospinal fluid. Clin Pharmacol Ther 2022; 111:826-834. [PMID: 35064573 PMCID: PMC9305158 DOI: 10.1002/cpt.2531] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/04/2022] [Indexed: 11/14/2022]
Abstract
Delivery of biologics via cerebrospinal fluid (CSF) has demonstrated potential to access the tissues of the central nervous system (CNS) by circumventing the blood‐brain barrier and blood‐CSF barrier. Developing an effective CSF drug delivery strategy requires optimization of multiple parameters, including choice of CSF access point, delivery device technology, and delivery kinetics to achieve effective therapeutic concentrations in the target brain region, whereas also considering the biologic modality, mechanism of action, disease indication, and patient population. This review discusses key preclinical and clinical examples of CSF delivery for different biologic modalities (antibodies, nucleic acid‐based therapeutics, and gene therapy) to the brain via CSF or CNS access routes (intracerebroventricular, intrathecal‐cisterna magna, intrathecal‐lumbar, intraparenchymal, and intranasal), including the use of novel device technologies. This review also discusses quantitative models of CSF flow that provide insight into the effect of fluid dynamics in CSF on drug delivery and CNS distribution. Such models can facilitate delivery device design and pharmacokinetic/pharmacodynamic translation from preclinical species to humans in order to optimize CSF drug delivery to brain regions of interest.
Collapse
Affiliation(s)
- Shraddha S Sadekar
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Mayumi Bowen
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Hao Cai
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Samira Jamalian
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Hanine Rafidi
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Whitney Shatz‐Binder
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Julien Lafrance‐Vanasse
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Pamela Chan
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - William J. Meilandt
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Amy Oldendorp
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Alavattam Sreedhara
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Ann Daugherty
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Susan Crowell
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Kristin R. Wildsmith
- Clinical pharmacology and translational medicine Neurology business Eisai, Nutley NJ 07110 USA
| | - Jasvinder Atwal
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Reina N. Fuji
- Genentech Research and Early Development Genentech, Inc., a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| | - Josh Horvath
- Pharma Technical Development. Genentech, Inc, a member of the Roche Group 1 DNA Way South San Francisco CA 94080 USA
| |
Collapse
|
14
|
McNeer JL, Schmiegelow K. Management of CNS Disease in Pediatric Acute Lymphoblastic Leukemia. Curr Hematol Malig Rep 2022; 17:1-14. [PMID: 35025035 DOI: 10.1007/s11899-021-00640-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The treatment of acute lymphoblastic leukemia (ALL) is one of the success stories of pediatric oncology, but challenges and questions remain, including the optimal approach to the treatment of central nervous system (CNS) leukemia. It is unclear why some children with ALL develop CNS leukemia and others do not, and there remains debate regarding optimal regimens for prophylaxis, upfront treatment, and the treatment of CNS relapses. These topics are especially important since both cranial radiation therapy (CRT) and intensive intrathecal therapy carry risks of both short- and long-term adverse effects. In this review, we aim to identify areas of ongoing debate on this topic, review the biology of CNS leukemia, and summarize clinical trial data that address some of these questions. RECENT FINDINGS Both retrospective and meta-analyses have demonstrated that few patients with ALL benefit from CRT as a component of CNS-directed treatment for de novo disease, allowing cooperative groups to greatly limit the number of patients undergoing CRT as part of their initial ALL regimens. More recent efforts are focusing on how best to assay for low levels of CNS disease at the time of diagnosis, as well as the biological drivers that may result in CNS leukemia in certain patients. Progress remains to be made in the identification and treatment of CNS leukemia in pediatric ALL. Advancements have occurred to limit the number of children undergoing CRT, but much has yet to be learned to better understand the biology of and risk factors for CNS leukemia, and novel approaches are required to approach CNS relapse of ALL.
Collapse
Affiliation(s)
- Jennifer L McNeer
- Section of Pediatric Hematology/Oncology/Stem Cell Transplant, University of Chicago Comer Children's Hospital, 5841 S. Maryland Ave, MC 4060, Chicago, IL, 60637, USA.
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
15
|
Craven CL, Gissen P, Bower R, Lee L, Aquilina K, Thompson DNP. A survival analysis of ventricular access devices for delivery of cerliponase alfa. J Neurosurg Pediatr 2022; 29:115-121. [PMID: 34624852 DOI: 10.3171/2021.7.peds21129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/08/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Late infantile neuronal ceroid lipofuscinosis type 2 (CLN2) is a rare autosomal recessive disease caused by tripeptidyl peptidase 1 enzyme deficiency. At the authors' center, the medication cerliponase alfa is administered every 2 weeks via the intracerebroventricular (ICV) route. This requires the placement of a ventricular access device (VAD) or reservoir and frequent percutaneous punctures of this device over the child's lifetime. In this study, the authors audited the longevity and survival of these VADs and examined the causes of device failure. METHODS A single-center survival analysis of VAD insertions and revisions (January 2014 through June 2020) was conducted. All children received cerliponase alfa infusions through a VAD. Patient characteristics and complications were determined from a prospectively maintained surgical database and patient records. For the VAD survival analysis, the defined endpoint was when the device was removed or changed. Reservoir survival was assessed using Kaplan-Meier curves and the log-rank (Cox-Mantel) test. RESULTS A total of 17 patients had VADs inserted for drug delivery; median (range) age at first surgery was 4 years 4 months (1 year 8 months to 15 years). Twenty-six VAD operations (17 primary insertions and 9 revisions) were required among these 17 patients. Twelve VAD operations had an associated complication, including CSF infection (n = 6) with Propionibacterium and Staphylococcus species being the most prevalent organisms, significant surgical site swelling preventing infusion (n = 3), leakage/wound breakdown (n = 2), and catheter obstruction (n = 1). There were no complications or deaths associated with VAD insertion. The median (interquartile range) number of punctures was 59.5 (7.5-82.0) for unrevised VADs (n = 17) versus 2 (6-87.5) for revised VADs (n = 9) (p = 0.70). The median survival was 301 days for revisional reservoirs (n = 9) versus 2317 days for primary inserted reservoirs (n = 17) (p = 0.019). CONCLUSIONS In the context of the current interest in intrathecal drug delivery for rare metabolic disorders, the need for VADs is likely to increase. Auditing the medium- to long-term outcomes associated with these devices will hopefully result in their wider application and may have potential implications on the development of new VAD technologies. These results could also be used to counsel parents prior to commencement of therapy and VAD implantation.
Collapse
Affiliation(s)
- Claudia L Craven
- 1Department of Neurosurgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Paul Gissen
- 1Department of Neurosurgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom.,2Department of Paediatric Metabolic Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom; and.,3UCL Institute of Child Health, London, United Kingdom
| | - Rebecca Bower
- 2Department of Paediatric Metabolic Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom; and
| | - Laura Lee
- 2Department of Paediatric Metabolic Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom; and
| | - Kristian Aquilina
- 1Department of Neurosurgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom.,3UCL Institute of Child Health, London, United Kingdom
| | - Dominic N P Thompson
- 1Department of Neurosurgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom.,3UCL Institute of Child Health, London, United Kingdom
| |
Collapse
|
16
|
Abed El Rahman SKED, Elshafy SSA, Samra M, Ali HM, Mohamed RA. PIM2 and NF-κβ gene expression in a sample of AML and ALL Egyptian patients and its relevance to response to treatment. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00162-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The relation between PIM2 and the transcriptional factor NF κβ have been controversial in literature. The significance of PIM2 and NF-κβ genes expression on the incidence of acute leukemia (AML and ALL) and its relevance to the response rate was evaluated. Sixty de novo acute leukemia patients were stratified in 2 groups: 30 acute myeloid leukemia (AML) and 30 acute lymphoblastic leukemia (ALL) patients and compared to 30 sex- and age-matched controls. The expression level of PIM2 and NF κβ genes was measured using quantitative real-time polymerase chain reaction (QRT-PCR). The patients were followed with clinical examination and complete blood counts.
Results
The expression level of PIM2 gene was significantly higher in AML patients (P<0.001) compared to the control group. The mean expression level of NF κβ gene was significantly high in AML and ALL patients compared to the healthy control group (P=0.037 and P<0.001; respectively). The overall survival in AML patients was higher in NF κβ gene low expressers compared to high expressers (P=0.047). The number of AML patients who achieved complete remission was significantly higher in PIM2 gene low expressers in comparison to PIM2 gene high expressers (P=0.042).
Conclusion
PIM2 and NF κβ genes might have a role in the pathogenesis of acute leukemia, poor overall survival, and failure of response to induction therapy.
Collapse
|
17
|
A phase 1/2 study of thiotepa-based immunochemotherapy in relapsed/refractory primary CNS lymphoma: the TIER trial. Blood Adv 2021; 5:4073-4082. [PMID: 34464973 PMCID: PMC8945638 DOI: 10.1182/bloodadvances.2021004779] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/07/2021] [Indexed: 11/25/2022] Open
Abstract
Thiotepa at 50 mg/m2 was safely incorporated into TIER immunochemotherapy. Despite a clinically meaningful treatment response rate, long-term survival was seen only with ASCT consolidation.
Relapsed or refractory primary central nervous system lymphoma (rrPCNSL) confers a poor prognosis with no accepted standard of care. Very few prospective studies have been conducted in this patient group. This study was a multicenter phase 1/2 study that investigated thiotepa in combination with ifosfamide, etoposide, and rituximab (TIER) for the treatment of PCNSL relapsed or refractory to high-dose methotrexate-based chemotherapy. A 3 + 3 design investigated the recommended phase 2 dose of thiotepa for a single-stage phase 2 cohort by assessing the activity of 2 cycles of TIER against rrPCNSL. The primary outcome was overall response rate. The dose-finding study demonstrated that 50 mg/m2 of thiotepa could be safely delivered within the TIER regimen. No dose-limiting toxicities were encountered in phase 1, and TIER was well-tolerated by the 27 patients treated in phase 2. The most common grade 3 to 4 toxicities were neutropenia (56% of patients) and thrombocytopenia (39%). An overall response was confirmed in 14 patients (52%), which met the prespecified threshold for clinically relevant activity. The median progression-free survival was 3 months (95% confidence interval [CI], 2 to 6 months) and overall survival 5 months (95% CI, 3 to 9 months). Exploratory analyses suggest a greater benefit for thiotepa-naïve patients. Six patients successfully completed autologous stem cell transplantation (ASCT) consolidation, with 4 experiencing durable remissions after a median follow-up of 50 months. The TIER regimen can be delivered safely and is active against rrPCNSL. When it is followed by ASCT, it can provide durable remission and long-term survival. However, for the majority of patients, prognosis remains poor, and novel treatment strategies are urgently needed. This trial was registered at https://www.clinicaltrialsregister.eu/ctr-search/search as EudraCT 2014-000227-24 and ISRCTN 12857473.
Collapse
|
18
|
Alcantara M, Fuentealba J, Soussain C. Emerging Landscape of Immunotherapy for Primary Central Nervous System Lymphoma. Cancers (Basel) 2021; 13:cancers13205061. [PMID: 34680209 PMCID: PMC8534133 DOI: 10.3390/cancers13205061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Primary central nervous system lymphoma (PCNSL) is characterized by its location in the central nervous system comprising the brain, the eye, the cerebrospinal fluid and the spinal cord and a poor prognosis with the current chemotherapies. Immunotherapies represent a new paradigm in the care of patients with B-cell lymphoma, but, till recently, immunotherapies studies excluded patients with PCNSL because of the lack of knowledge on the immune network in the brain. Recent studies shed a new light on the origin and characteristics of the CNS immune cells. We review the current experimental preclinical and clinical developments of immunotherapies in CNS lymphoma as well as the effects of targeted therapies on the brain microenvironment. We provide perspectives for improving the efficacy of immunotherapies in the specific setting of PCNSL for a better prognosis of this disease. Abstract Primary central nervous system lymphoma (PCNSL) is, mainly, a diffuse large B-cell lymphoma (DLBCL) with a non-germinal center B-cell (non-GCB) origin. It is associated with a poor prognosis and an unmet medical need. Immunotherapy has emerged as one of the most promising areas of research and is now part of the standard treatment for many solid and hematologic tumors. This new class of therapy generated great enthusiasm for the treatment of relapsed/refractory PCNSL. Here, we discuss the challenges of immunotherapy for PCNSL represented by the lymphoma cell itself and the specific immune brain microenvironment. We review the current clinical development from the anti-CD20 monoclonal antibody to CAR-T cells, as well as immune checkpoint inhibitors and targeted therapies with off-tumor effects on the brain microenvironment. Perspectives for improving the efficacy of immunotherapies and optimizing their therapeutic role in PCNSL are suggested.
Collapse
Affiliation(s)
- Marion Alcantara
- Center for Cancer Immunotherapy, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France; (M.A.); (J.F.)
- Clinical Hematology Unit, Institut Curie, 92210 Saint-Cloud, France
| | - Jaime Fuentealba
- Center for Cancer Immunotherapy, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France; (M.A.); (J.F.)
| | - Carole Soussain
- Center for Cancer Immunotherapy, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France; (M.A.); (J.F.)
- Clinical Hematology Unit, Institut Curie, 92210 Saint-Cloud, France
- Correspondence:
| |
Collapse
|
19
|
Ambady P, Doolittle ND, Fox CP. Relapsed and refractory primary CNS lymphoma: treatment approaches in routine practice. ANNALS OF LYMPHOMA 2021; 5:23. [PMID: 35253010 PMCID: PMC7612457 DOI: 10.21037/aol-21-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Despite recent therapeutic progress and improved survival for many patients with primary central nervous system lymphoma (PCNSL), up to 50% of patients will experience refractory or relapsed disease following first-line treatment with high dose methotrexate (HD-MTX) based regimens. The majority of such events occur within 2 years of diagnosis although, unlike their systemic counterpart, the risk of PCNSL relapse remains, even for patients in radiologic complete response at 10 years following diagnosis. Currently, there are no approved therapies, and no widely accepted 'standard-of-care' approaches for the treatment of refractory or recurrent primary central nervous system lymphoma (rrPCNSL). Re-treatment with HD-MTX based regimens, use of non-cross resistant chemotherapy regimens, high-dose chemotherapy and autologous stem cell transplantation (HDT-ASCT), and brain irradiation all remain important therapeutic approaches for rrPCNSL. However, the survival outcomes for patients with rrPCNSL remain extremely poor and the vast majority of patients will die of their disease. Increasingly, novel treatment approaches are being investigated in early phase clinical studies. Importantly, such therapies need to be evaluated in the context of both refractory and relapsed disease; in older patients and those with co-morbid conditions; and those with neurocognitive dysfunction. A deeper understanding of the molecular genetic mechanisms underpinning rrPCNSL and its unique tumor microenvironment is urgently needed to inform biologically rational and effective therapies. rrPCNSL remains a clear unmet clinical need and a high priority area for clinical research that will require national and international collaborative studies with embedded translational science in order to improve outcomes for patients.
Collapse
Affiliation(s)
- Prakash Ambady
- Department of Neurology, Neuro-Oncology and Blood Brain Barrier Program, Oregon Health & Science University, Portland, Oregon, USA
| | - Nancy D. Doolittle
- Department of Neurology, Neuro-Oncology and Blood Brain Barrier Program, Oregon Health & Science University, Portland, Oregon, USA
| | - Christopher P. Fox
- Department of Clinical Haematology, Nottingham University Hospitals NHS Trust, Nottingham, UK
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
20
|
Wen J, Cheever T, Wang L, Wu D, Reed J, Mascola J, Chen X, Liu C, Pegu A, Sacha JB, Lu Y, Haigwood NL, Chen ISY. Improved delivery of broadly neutralizing antibodies by nanocapsules suppresses SHIV infection in the CNS of infant rhesus macaques. PLoS Pathog 2021; 17:e1009738. [PMID: 34283885 PMCID: PMC8323878 DOI: 10.1371/journal.ppat.1009738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/30/2021] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) directed to HIV-1 have shown promise at suppressing viremia in animal models. However, the use of bNAbs for the central nervous system (CNS) infection is confounded by poor penetration of the blood brain barrier (BBB). Typically, antibody concentrations in the CNS are extremely low; with levels in cerebrospinal fluid (CSF) only 0.1% of blood concentrations. Using a novel nanotechnology platform, which we term nanocapsules, we show effective transportation of the human bNAb PGT121 across the BBB in infant rhesus macaques upon systemic administration up to 1.6% of plasma concentration. We demonstrate that a single dose of PGT121 encased in nanocapsules when delivered at 48h post-infection delays early acute infection with SHIVSF162P3 in infants, with one of four animals demonstrating viral clearance. Importantly, the nanocapsule delivery of PGT121 improves suppression of SHIV infection in the CNS relative to controls. In patients where HIV-1 is fully suppressed by antiretroviral drugs, HIV-1 still persists in reservoirs. If antiretroviral drugs are stopped, the virus will emerge from these reservoirs and re-seeds systemically. The central nervous system (CNS) is proposed to be a tissue compartment that harbors other HIV-1 reservoirs. A key obstacle that constrains the treatment for the CNS infection is the blood–brain barrier (BBB), a highly restrictive barrier separating the circulating blood from the brain and extracellular fluid in the CNS, which impedes ~98% of the small molecule therapeutics and almost all macromolecules including broadly neutralizing antibodies (bNAbs) directed to HIV-1. Our “nanocapsule” strategy is based on a nanotechnology wherein bNAb molecules are encapsulated within nanocapsules of which the surface contains abundant choline and acetylcholine analogues. This design allows the nanocapsules to effectively cross the BBB to deliver bNAbs into the CNS upon systemic administration and show an impact of bNAb on CNS reservoirs in SHIV infected infant macaques.
Collapse
Affiliation(s)
- Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), UCLA AIDS Institute, Los Angeles, California, United States of America
| | - Tracy Cheever
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Lan Wang
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), UCLA AIDS Institute, Los Angeles, California, United States of America
| | - Di Wu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, California, United States of America
| | - Jason Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - John Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Xuejun Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Cuiping Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America.,Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, California, United States of America
| | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Irvin S Y Chen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), UCLA AIDS Institute, Los Angeles, California, United States of America
| |
Collapse
|
21
|
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: 32] [Impact Index Per Article: 10.7] [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
|
22
|
|
23
|
No Early Effect of Intrathecal Rituximab in Progressive Multiple Sclerosis (EFFRITE Clinical Trial). Mult Scler Int 2021; 2021:8813498. [PMID: 33763241 PMCID: PMC7964121 DOI: 10.1155/2021/8813498] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/22/2021] [Accepted: 02/27/2021] [Indexed: 11/30/2022] Open
Abstract
Background The progressive phase of multiple sclerosis (MS) is characterized by an intrathecal (IT) compartmentalization of inflammation, involving B-cells within meningeal follicles, and resisting all the available immunosuppressive treatments. A new therapeutic paradigm may be to target this inflammation by injecting immunosuppressive drugs inside the central nervous system compartment. Methods We designed a single-center, open-label, randomized, controlled, phase II study designed to evaluate the safety and efficacy of IT rituximab in progressive MS (EFFRITE trial; ClinicalTrial Registration NCT02545959). Patients were randomized into three arms (1 : 1 : 1): control group, IT rituximab (20 mg, IT) group, and intravenous+IT (IV+IT) group. The main outcome was a change in levels of CSF biomarkers of inflammation (osteopontin). Secondary outcomes were changes in levels of CSF biomarkers of axonal loss (neurofilament light chain) and clinical and MRI changes. Results Ten patients were included (2 : 4 : 4). No adverse event occurred. OPN level remained stable in CSF at each time point, whereas NFL had slightly decreased (-8.7%) at day 21 (p = 0.02). Clinical parameters remained stable and leptomeningeal enhancements remained unchanged. Conclusion Clinical outcome and biomarkers of inflammation were not dramatically modified after IT injection of rituximab, probably due to its limited efficiency in CSF. Drug issues for future studies are discussed.
Collapse
|
24
|
Schaff LR, Ambady P, Doolittle ND, Grommes C. Primary central nervous system lymphoma: a narrative review of ongoing clinical trials and goals for future studies. ACTA ACUST UNITED AC 2021; 5. [PMID: 33912868 PMCID: PMC8078860 DOI: 10.21037/aol-20-47] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare disease of the brain, spine, cerebrospinal fluid (CSF) and/or vitreoretinal space. PCNSL is chemo and radiosensitive but relapse is common even years after initial treatment. Outside of consensus regarding the use of high-dose methotrexate (HD-MTX) for first line treatment, there is little uniformity in the management of newly diagnosed or relapsed PCNSL. The lack of consensus is driven by a paucity of randomized trials in this disease. Prospective studies are troubled by low enrollment, the lack of a standard induction regimen, and a varied approach to consolidation strategies. Moreover, the PCNSL patient population is heterogeneous and includes a high proportion of elderly or frail patients and consists of patients manifesting disease in varied compartments of the central nervous system (CNS). As a result, current treatment strategies vary widely and are often dictated by physician and institutional preference or regional practice. This review provides an overview of recently completed and ongoing therapeutic studies for patients with newly diagnosed and recurrent or refractory PCNSL. It discusses the existing evidence behind common approaches to induction and consolidation or maintenance regimens as well as the recent data regarding management of recurrent disease. Finally, it highlights the complexity of trial design in this disease and provides a framework for the design of future studies, which are needed to identify patient populations likely to benefit from specific induction, consolidation, or maintenance therapies.
Collapse
Affiliation(s)
- Lauren R Schaff
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prakash Ambady
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Nancy D Doolittle
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Christian Grommes
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
25
|
Singh M, Singh SP, Yadav D, Agarwal M, Agarwal S, Agarwal V, Swargiary G, Srivastava S, Tyagi S, Kaur R, Mani S. Targeted Delivery for Neurodegenerative Disorders Using Gene Therapy Vectors: Gene Next Therapeutic Goals. Curr Gene Ther 2021; 21:23-42. [PMID: 32811395 DOI: 10.2174/1566523220999200817164907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 11/22/2022]
Abstract
The technique of gene therapy, ever since its advent nearly fifty years ago, has been utilized by scientists as a potential treatment option for various disorders. This review discusses some of the major neurodegenerative diseases (NDDs) like Alzheimer's disease (AD), Parkinson's Disease (PD), Motor neuron diseases (MND), Spinal Muscular Atrophy (SMA), Huntington's Disease (HD), Multiple Sclerosis (MS), etc. and their underlying genetic mechanisms along with the role that gene therapy can play in combating them. The pathogenesis and the molecular mechanisms specifying the altered gene expression of each of these NDDs have also been discussed in elaboration. The use of gene therapy vectors can prove to be an effective tool in the field of curative modern medicine for the generations to come. Therefore, consistent efforts and progressive research towards its implementation can provide us with powerful treatment options for disease conditions that have so far been considered as incurable.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P, India
| | - Surinder P Singh
- Bhartiya Nirdeshak Dravya Division, CSIR-National Physical Laboratory, New Delhi, India
| | - Deepshikha Yadav
- Bhartiya Nirdeshak Dravya Division, CSIR-National Physical Laboratory, New Delhi, India
| | - Mugdha Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Shriya Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Geeta Swargiary
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Sahil Srivastava
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Sakshi Tyagi
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Ramneek Kaur
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| |
Collapse
|
26
|
Negron A, Stüve O, Forsthuber TG. Ectopic Lymphoid Follicles in Multiple Sclerosis: Centers for Disease Control? Front Neurol 2020; 11:607766. [PMID: 33363512 PMCID: PMC7753025 DOI: 10.3389/fneur.2020.607766] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
While the contribution of autoreactive CD4+ T cells to the pathogenesis of Multiple Sclerosis (MS) is widely accepted, the advent of B cell-depleting monoclonal antibody (mAb) therapies has shed new light on the complex cellular mechanisms underlying MS pathogenesis. Evidence supports the involvement of B cells in both antibody-dependent and -independent capacities. T cell-dependent B cell responses originate and take shape in germinal centers (GCs), specialized microenvironments that regulate B cell activation and subsequent differentiation into antibody-secreting cells (ASCs) or memory B cells, a process for which CD4+ T cells, namely follicular T helper (TFH) cells, are indispensable. ASCs carry out their effector function primarily via secreted Ig but also through the secretion of both pro- and anti-inflammatory cytokines. Memory B cells, in addition to being capable of rapidly differentiating into ASCs, can function as potent antigen-presenting cells (APCs) to cognate memory CD4+ T cells. Aberrant B cell responses are prevented, at least in part, by follicular regulatory T (TFR) cells, which are key suppressors of GC-derived autoreactive B cell responses through the expression of inhibitory receptors and cytokines, such as CTLA4 and IL-10, respectively. Therefore, GCs represent a critical site of peripheral B cell tolerance, and their dysregulation has been implicated in the pathogenesis of several autoimmune diseases. In MS patients, the presence of GC-like leptomeningeal ectopic lymphoid follicles (eLFs) has prompted their investigation as potential sources of pathogenic B and T cell responses. This hypothesis is supported by elevated levels of CXCL13 and circulating TFH cells in the cerebrospinal fluid (CSF) of MS patients, both of which are required to initiate and maintain GC reactions. Additionally, eLFs in post-mortem MS patient samples are notably devoid of TFR cells. The ability of GCs to generate and perpetuate, but also regulate autoreactive B and T cell responses driving MS pathology makes them an attractive target for therapeutic intervention. In this review, we will summarize the evidence from both humans and animal models supporting B cells as drivers of MS, the role of GC-like eLFs in the pathogenesis of MS, and mechanisms controlling GC-derived autoreactive B cell responses in MS.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Neurology Section, Veterans Affairs North Texas Health Care System, Medical Service, Dallas, TX, United States
| | - Thomas G Forsthuber
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
27
|
Wang X, Huynh C, Urak R, Weng L, Walter M, Lim L, Vyas V, Chang WC, Aguilar B, Brito A, Sarkissian A, Bandara NA, Yang L, Wang J, Wu X, Zhang J, Priceman SJ, Qin H, Kwak LW, Budde LE, Thomas SH, Clark MC, Popplewell L, Siddiqi T, Brown CE, Forman SJ. The Cerebroventricular Environment Modifies CAR T Cells for Potent Activity against Both Central Nervous System and Systemic Lymphoma. Cancer Immunol Res 2020; 9:75-88. [PMID: 33093217 DOI: 10.1158/2326-6066.cir-20-0236] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/24/2020] [Accepted: 10/15/2020] [Indexed: 11/16/2022]
Abstract
Lymphomas with central nervous system (CNS) involvement confer a worse prognosis than those without CNS involvement, and patients currently have limited treatment options. T cells genetically engineered with CD19-targeted chimeric antigen receptors (CAR) are effective against B-cell malignancies and show tremendous potential in the treatment of systemic lymphoma. We aimed to leverage this strategy toward a more effective therapy for patients with lymphoma with CNS disease. NOD-scid IL2Rgammanull (NSG) mice with CNS and/or systemic lymphoma were treated with CD19-CAR T cells via intracerebroventricular (ICV) or intravenous (IV) injection. CAR T cells isolated after treatment were rigorously examined for phenotype, gene expression, and function. We observed that CAR T cells infused ICV, but not IV, completely and durably eradicated both CNS and systemic lymphoma. CAR T cells delivered ICV migrated efficiently to the periphery, homed to systemic tumors, and expanded in vivo, leading to complete elimination of disease and resistance to tumor rechallenge. Mechanistic studies indicated that ICV-delivered CAR T cells are conditioned by exposure to cerebrospinal fluid in the ICV environment for superior antilymphoma activity and memory function compared with IV-delivered CAR T cells. Further analysis suggested that manipulating cellular metabolism or preactivating therapeutic CAR T cells with antigen ex vivo may improve the efficacy of CAR T cells in vivo Our demonstration that ICV-delivered CD19-CAR T cells had activity against CNS and systemic lymphoma could offer a valuable new strategy for treatment of B-cell malignancies with CNS involvement.
Collapse
Affiliation(s)
- Xiuli Wang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California.
| | - Christian Huynh
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Ryan Urak
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Lihong Weng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Miriam Walter
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Laura Lim
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Vibhuti Vyas
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Wen-Chung Chang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Brenda Aguilar
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Alfonso Brito
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Aniee Sarkissian
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - N Achini Bandara
- Clinical and Translational Project Development Core, City of Hope, Duarte, California
| | - Lu Yang
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, California
| | - Jinhui Wang
- Integrative Genomics Core, Beckman Research Institute, City of Hope, Duarte, California
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute, City of Hope, Duarte, California
| | - Jianying Zhang
- The Department of Computational and Quantitative Medicine, City of Hope, Duarte, California
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Hong Qin
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope, Duarte, California
| | - Larry W Kwak
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope, Duarte, California
| | - Lihua E Budde
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Sandra H Thomas
- Clinical and Translational Project Development Core, City of Hope, Duarte, California
| | - Mary C Clark
- Clinical and Translational Project Development Core, City of Hope, Duarte, California
| | - Leslie Popplewell
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Tanya Siddiqi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Christine E Brown
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| |
Collapse
|
28
|
Abstract
New evidence and increased use of intracranial devices have increased the frequency of intraventricular (IVT) medication administration in the neurologic intensive care unit. Significant benefits and risks are associated with administration of medications directly into the central nervous system. This review summarizes important literature, along with key information for clinicians regarding the administration, dosing, monitoring, and adverse effects related to IVT medication usage. Multiple medications have supporting literature for their use in critically ill patients including amphotericin B, aminoglycosides, colistimethate, daptomycin, quinupristin/dalfopristin, vancomycin, alteplase, and nicardipine. Sterile preparation and delivery, along with different types of devices that support medication administration, are also reviewed. One randomized, placebo-controlled trial of alteplase demonstrated decreased mortality but no change in good functional outcome. Other reports of IVT medication use are mainly limited to case reports and retrospective case series. There is a need for increased research on the topic; however, several practical barriers decrease the likelihood of a large, placebo-controlled, prospective study for most indications. Providers should consider implementing protocols to maximize safety of IVT medication delivery to ensure optimal patient outcomes.
Collapse
|
29
|
Tsang M, Cleveland J, Rubenstein JL. On point in primary CNS lymphoma. Hematol Oncol 2020; 38:640-647. [PMID: 32510610 DOI: 10.1002/hon.2761] [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: 05/09/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 01/03/2023]
Abstract
Primary CNS lymphoma (PCNSL) is an aggressive brain tumor that represents a significant challenge both to elucidate its biological pathogenesis as well as to develop definitive precision medicines with minimal collateral toxicity. We highlight the key issues in diagnosis and treatment and focus on emerging technologies, current options among consolidation strategies, and biological agents. We anticipate that further development of molecular diagnostics and molecular imaging approaches that elucidate minimal residual disease in brain parenchyma, leptomeninges, intraocular compartments and even bone marrow will greatly impact the delivery and timing of cytotoxic and biological therapies. Implementation of these approaches is likely essential to clarify ongoing discrepancies in the interpretation of clinical trial results that currently are based on relatively unrefined definitions of response. While the results of early phase investigations involving ibrutinib and the IMiD agents, lenalidomide, pomalidomide, as well as avadomide, strongly support the hypothesis that the B-cell receptor (BCR) pathway, involving MYD88 and CD79B and NF-kB activation, is critical to the pathogenesis of PCNSL, much work is needed to elucidate mechanisms of resistance. Similarly, development of strategies to overcome immunosuppressive mechanisms that are upregulated in the tumor microenvironment is a high priority. Finally, ongoing evidence supports the hypothesis that the blood-brain barrier represents a significant impediment to efficient brain tumor penetration of novel therapeutic agents and innovative strategies of drug delivery remain essential to further improve outcomes.
Collapse
Affiliation(s)
- Mazie Tsang
- Division of Hematology/Oncology, University of California, San Francisco, California, USA
| | - Joseph Cleveland
- Department of Medicine, University of California, San Francisco, California, USA
| | - James L Rubenstein
- Division of Hematology/Oncology, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| |
Collapse
|
30
|
Intraventricular Rituximab in Pediatric CD20-positive Refractory Primary Central Nervous System Lymphoma. J Pediatr Hematol Oncol 2019; 41:571-573. [PMID: 30124546 DOI: 10.1097/mph.0000000000001291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and aggressive type of extranodal non-Hodgkin lymphoma that carries an unsatisfactory prognosis. Treating refractory PCNSL is challenging because of resistance to conventional cytotoxic and intrathecal chemotherapies. Therefore, novel therapeutic approaches are needed. Here, we report a 12-year-old boy with CD20-positive PCNSL, which was refractory to combination chemotherapy and intravenous rituximab. However, the patient achieved complete remission after repeated intraventricular rituximab administration. The results of this case indicate that intraventricular rituximab is an effective option to treat refractory PCNSL in children.
Collapse
|
31
|
Abstract
OBJECTIVES Anti-N-methyl-D-aspartate receptor encephalitis is considered an immune-mediated form of encephalitis with paraneoplastic and nonparaneoplastic forms. Delay in recognition is common and patients typically present to the ICU without a diagnosis or with complications following a delayed diagnosis. The aim of this review is to provide a focused overview for the ICU clinician regarding presentation, diagnosis, and critical care management. DATA SOURCES, STUDY SELECTION, AND DATA EXTRACTION PubMed database search with manual review of articles involving anti-N-methyl-D-aspartate receptor encephalitis. DATA SYNTHESIS Anti-N-methyl-D-aspartate receptor encephalitis is increasingly encountered in the ICU. The cascade of events initiating anti-N-methyl-D-aspartate receptor antibody formation may involve an infectious trigger particularly in the setting of teratoma. Following a prodrome, most patients develop psychiatric symptoms followed by movement disorder. Classical, psychiatric, and catatonic phenotypes may be distinguished based on the presence and severity of symptoms. Early immunotherapy and low initial cerebrospinal fluid inflammation are independent predictors of positive outcomes in ICU patients. Concomitant organ failure, status epilepticus, and the identification of a tumor did not influence outcome in critically ill patients. Supportive care in the ICU includes management of various manifestations of dyskinesia, status epilepticus, autonomic disorders, and the need for general sedation. Common treatment strategies and limitations are discussed including the emerging role of bortezomib. CONCLUSIONS Intensivists should be familiar with the presentation and management of anti-N-methyl-D-aspartate receptor encephalitis. Early diagnosis and immediate implementation of steroids, immunoglobulins, and/or plasmapheresis and immune therapy are associated with a good neurologic outcome although response may be delayed. The selection and timing of second-line immune therapy requires further study.
Collapse
|
32
|
Mondello P, Mian M, Bertoni F. Primary central nervous system lymphoma: Novel precision therapies. Crit Rev Oncol Hematol 2019; 141:139-145. [DOI: 10.1016/j.critrevonc.2019.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/01/2023] Open
|
33
|
Sustained delivery and molecular targeting of a therapeutic monoclonal antibody to metastases in the central nervous system of mice. Nat Biomed Eng 2019; 3:706-716. [PMID: 31384008 PMCID: PMC6736720 DOI: 10.1038/s41551-019-0434-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/26/2019] [Indexed: 11/24/2022]
Abstract
Approximately 15–40% of all cancers develop metastases in the
central nervous system (CNS), yet few therapeutic options exist to treat them.
Cancer therapies based on monoclonal antibodies are widely successful, yet have
limited efficacy against CNS metastases, owing to the low levels of the drug
reaching the tumour site. Here, we show that the encapsulation of rituximab
within a crosslinked zwitterionic polymer layer leads to the sustained release
of rituximab as the crosslinkers are gradually hydrolyzed, enhancing by
approximately 10-fold the CNS levels of the antibody with respect to the
administration of naked rituximab. When the nanocapsules are functionalized with
CXCL13, the ligand for the chemokine receptor CXCR5 frequently found on B-cell
lymphoma, a single dose led to improved control of CXCR5-expressing metastases
in a murine xenograft model of non-Hodgkin lymphoma, and eliminated lymphoma in
a xenografted humanized bone-marrow–liver–thymus mouse model.
Encapsulation and molecular targeting of therapeutic antibodies could become an
option for the treatment of cancers with CNS metastases.
Collapse
|
34
|
Secondary Diffuse Choroid Plexus B-Cell Lymphoma: Case Report and Review of Literature. World Neurosurg 2019; 128:18-22. [DOI: 10.1016/j.wneu.2019.04.203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/21/2019] [Accepted: 04/22/2019] [Indexed: 11/20/2022]
|
35
|
Abdul Razzak R, Florence GJ, Gunn-Moore FJ. Approaches to CNS Drug Delivery with a Focus on Transporter-Mediated Transcytosis. Int J Mol Sci 2019; 20:E3108. [PMID: 31242683 PMCID: PMC6627589 DOI: 10.3390/ijms20123108] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/15/2019] [Accepted: 06/16/2019] [Indexed: 12/13/2022] Open
Abstract
Drug delivery to the central nervous system (CNS) conferred by brain barriers is a major obstacle in the development of effective neurotherapeutics. In this review, a classification of current approaches of clinical or investigational importance for the delivery of therapeutics to the CNS is presented. This classification includes the use of formulations administered systemically that can elicit transcytosis-mediated transport by interacting with transporters expressed by transvascular endothelial cells. Neurotherapeutics can also be delivered to the CNS by means of surgical intervention using specialized catheters or implantable reservoirs. Strategies for delivering drugs to the CNS have evolved tremendously during the last two decades, yet, some factors can affect the quality of data generated in preclinical investigation, which can hamper the extension of the applications of these strategies into clinically useful tools. Here, we disclose some of these factors and propose some solutions that may prove valuable at bridging the gap between preclinical findings and clinical trials.
Collapse
Affiliation(s)
- Rana Abdul Razzak
- Medical and Biological Sciences Building, School of Biology, University of St Andrews, St Andrews KY16 9TF, UK.
- Biomedical Science Research Centre, Schools of Chemistry and Biology, University of St Andrews, St Andrews KY16 9TF, UK.
| | - Gordon J Florence
- Biomedical Science Research Centre, Schools of Chemistry and Biology, University of St Andrews, St Andrews KY16 9TF, UK.
| | - Frank J Gunn-Moore
- Medical and Biological Sciences Building, School of Biology, University of St Andrews, St Andrews KY16 9TF, UK.
- Biomedical Science Research Centre, Schools of Chemistry and Biology, University of St Andrews, St Andrews KY16 9TF, UK.
| |
Collapse
|
36
|
Phase 1 investigation of lenalidomide/rituximab plus outcomes of lenalidomide maintenance in relapsed CNS lymphoma. Blood Adv 2019; 2:1595-1607. [PMID: 29986852 DOI: 10.1182/bloodadvances.2017014845] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 05/08/2018] [Indexed: 12/29/2022] Open
Abstract
There is an unmet need for effective biological therapies for relapsed central nervous system (CNS) lymphoma. Lenalidomide is active in activated B-cell type diffuse large B-cell lymphoma and rituximab is effective in CNS lymphoma. These observations are the basis for this first trial of an immunomodulatory drug as monotherapy in CNS lymphoma, and, in patients with inadequate responses to lenalidomide, with rituximab. In an independent cohort, we evaluated lenalidomide maintenance after salvage with high-dose methotrexate or focal irradiation in relapsed primary CNS lymphoma (PCNSL). We determined safety, efficacy, and cerebrospinal fluid (CSF) penetration of lenalidomide at 10-, 15-, and 20-mg dose levels in 14 patients with refractory CD20+ CNS lymphoma. Nine subjects with relapsed, refractory CNS lymphoma achieved better than partial response with lenalidomide monotherapy, 6 maintained response ≥9 months, and 4 maintained response ≥18 months. Median progression-free survival for lenalidomide/rituximab was 6 months. In the independent cohort, response duration with lenalidomide maintenance after complete responses 2 through 5 were significantly longer than response durations after standard therapy. The CSF/plasma partition coefficient of lenalidomide was ≥20% at 15- and 20-mg dose levels. Change in CSF interleukin-10 at 1 month correlated with clinical response and response duration to lenalidomide. Metabolomic profiling of CSF identified novel biomarkers, including lactate, and implicated indoleamine-2,3 dioxygenase activity with CNS lymphoma progression on lenalidomide. We conclude that lenalidomide penetrates ventricular CSF and is active as monotherapy in relapsed CNS lymphomas. We provide evidence that maintenance lenalidomide potentiates response duration after salvage in relapsed PCNSL and delays whole brain radiotherapy (WBRT). This trial was registered at www.clinicaltrials.gov as #NCT01542918.
Collapse
|
37
|
Grommes C, Nayak L, Tun HW, Batchelor TT. Introduction of novel agents in the treatment of primary CNS lymphoma. Neuro Oncol 2019; 21:306-313. [PMID: 30423172 PMCID: PMC6380407 DOI: 10.1093/neuonc/noy193] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Novel insights into the pathophysiology of primary central nervous system lymphoma (PCNSL) have identified the B-cell receptor and Toll-like receptor pathway as well as immune evasion and suppressed tumor immune microenvironment as a key mechanism in the pathogenesis of PCNSL. Small molecules and novel agents targeting these aberrant pathways have been introduced into clinical trials targeting the recurrent or refractory PCNSL patient population. Agents like the Bruton tyrosine kinase (BTK) inhibitor ibrutinib or immunomodulatory drugs (IMiDs) like pomalidomide and lenalidomide have shown promising high response rates in the salvage setting. Here, we give an overview about the recent, exciting developments in PCNSL and summarize the results of clinical trials using novel agents in the recurrent and refractory salvage setting, which include immune checkpoint inhibitors, IMiDs, as well as BTK, phosphatidylinositol-3 kinase, and mammalian target of rapamycin inhibitors.
Collapse
Affiliation(s)
- Christian Grommes
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lakshmi Nayak
- Center for NeuroOncology, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Han W Tun
- Department of Hematology and Oncology and Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Tracy T Batchelor
- Departments of Neurology and Radiation Oncology, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
38
|
Bhargava P, Wicken C, Smith MD, Strowd RE, Cortese I, Reich DS, Calabresi PA, Mowry EM. Trial of intrathecal rituximab in progressive multiple sclerosis patients with evidence of leptomeningeal contrast enhancement. Mult Scler Relat Disord 2019; 30:136-140. [PMID: 30771580 DOI: 10.1016/j.msard.2019.02.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/01/2019] [Accepted: 02/09/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Leptomeningeal inflammation is associated with increased cortical damage and worse clinical outcomes in MS. It may be detected on contrast-enhanced T2-FLAIR imaging as focal leptomeningeal contrast-enhancement (LME). OBJECTIVE To assess the safety of intrathecal (IT) rituximab in progressive MS (PMS) and to assess its effects on LME and CSF biomarkers. METHODS PMS patients had a screening MRI to detect LME. Participants satisfying eligibility criteria underwent two IT administrations of 25 mg rituximab 2 weeks apart. Follow-up lumbar puncture and MRI were performed at 8 and 24 weeks after the first treatment. RESULTS Of 36 patients screened 15 had LME, 11 consented, and 8 received study treatment. Mean age was 56.7 years and number of LME lesions ranged from 1 to 3. No serious adverse effects occurred. We noted profound reductions in peripheral B cells from baseline to week 2 and 8 with some return at week 24. We also observed transient reductions in CSF B cells and CXCL-13 levels with an increase in BAFF levels. However, the number of LME did not change following treatment. CONCLUSIONS IT rituximab was well tolerated in PMS patients and had transient effects on CSF biomarkers but did not change LME.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Pathology 627, Baltimore, MD 21287, USA.
| | - Cassie Wicken
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Pathology 627, Baltimore, MD 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Pathology 627, Baltimore, MD 21287, USA
| | - Roy E Strowd
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Pathology 627, Baltimore, MD 21287, USA
| | - Irene Cortese
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Pathology 627, Baltimore, MD 21287, USA
| | - Ellen M Mowry
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Pathology 627, Baltimore, MD 21287, USA
| |
Collapse
|
39
|
Cai Q, Fang Y, Young KH. Primary Central Nervous System Lymphoma: Molecular Pathogenesis and Advances in Treatment. Transl Oncol 2019; 12:523-538. [PMID: 30616219 PMCID: PMC6371000 DOI: 10.1016/j.tranon.2018.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/11/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a group of extranodal non-Hodgkin lymphoma that exhibits specific biological characteristics and clinical behavior, with an aggressive disease course and unsatisfactory patient outcomes. It is of great importance to identify aberrant genetic loci and important molecular pathways that might suggest potential targets for new therapeutics and provide prognostic information. In this review, we listed various genetic and epigenetic alterations that are involved in PCNSL pathogenesis. In the aspect of treatment, we summarized the related literatures and evaluated the efficacy of surgery, induction chemotherapy, radiotherapy, intrathecal chemotherapy, and autologous stem cell transplantation in PCNSL. We also proposed the possible new agents for recurrent and relapse PCNSL based on the result of recent clinical researches.
Collapse
Affiliation(s)
- Qingqing Cai
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, P.R China.
| | - Yu Fang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, P.R China
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
40
|
Wicken C, Nguyen J, Karna R, Bhargava P. Leptomeningeal inflammation in multiple sclerosis: Insights from animal and human studies. Mult Scler Relat Disord 2018; 26:173-182. [DOI: 10.1016/j.msard.2018.09.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/16/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022]
|
41
|
Slavc I, Cohen-Pfeffer JL, Gururangan S, Krauser J, Lim DA, Maldaun M, Schwering C, Shaywitz AJ, Westphal M. Best practices for the use of intracerebroventricular drug delivery devices. Mol Genet Metab 2018; 124:184-188. [PMID: 29793829 DOI: 10.1016/j.ymgme.2018.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 04/23/2018] [Accepted: 05/09/2018] [Indexed: 01/01/2023]
Abstract
For decades, intracerebroventricular (ICV), or intraventricular, devices have been used in the treatment of a broad range of pediatric and adult central nervous system (CNS) disorders. Due to the limited permeability of the blood brain barrier, diseases with CNS involvement may require direct administration of drugs into the brain to achieve full therapeutic effect. A recent comprehensive literature review on the clinical use and complications of ICV drug delivery revealed that device-associated complication rates are variable, and may be as high as 33% for non-infectious complications and 27% for infectious complications. The variability in reported safety outcomes may be driven by a lack of consensus on best practices of device use. Numerous studies have demonstrated that employing strict aseptic techniques and following stringent protocols can dramatically reduce complications. Key practices to be considered in facilitating the safe, long-term use of these devices are presented.
Collapse
Affiliation(s)
- Irene Slavc
- Medical University of Vienna, Department of Pediatrics and Adolescent Medicine, Vienna, Austria.
| | | | | | - Jeanne Krauser
- McKnight Brain Institute, University of Florida, Gainsville, FL, USA
| | - Daniel A Lim
- University of California, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
42
|
Mendez JS, Grommes C. Treatment of Primary Central Nervous System Lymphoma: From Chemotherapy to Small Molecules. Am Soc Clin Oncol Educ Book 2018; 38:604-615. [PMID: 30231317 DOI: 10.1200/edbk_200829] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare form of extranodal non-Hodgkin lymphoma that is typically confined to the brain, eyes, and cerebrospinal fluid (CSF) without evidence of systemic spread. PCNSL is an uncommon tumor, and only four randomized trials and one phase III trial have been completed so far, all in the first-line setting. The prognosis of patients with PCNSL has improved during the past few decades with the introduction of high-dose methotrexate (HD-MTX), which now serves as the backbone of all first-line treatment regimens. Despite recent progress, results after treatment are durable in half of patients, and therapy can be associated with late neurotoxicity. Novel insights into the pathophysiology of PCNSL have identified the B-cell receptor (BCR) pathway as a key mechanism in the pathogenesis of PCNSL. The use of novel agents targeting components of the BCR pathway, namely the Bruton tyrosine kinase (BTK) inhibitor ibrutinib, and immunomodulatory drugs (IMIDs) like lenalidomide and pomalidomide, has so far been limited to patients who have recurrent/refractory PCNSL with promising high response rates. Within the past 5 years, there has been a peak in clinical trials investigating small molecules and novel reagents in the recurrent/refractory setting, including immune checkpoint inhibitors, IMIDs, and BTK and PI3K/AKT/mTOR inhibitors.
Collapse
Affiliation(s)
- Joe S Mendez
- From the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Neurology, Weill Cornell Medical College, New York, NY
| | - Christian Grommes
- From the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Neurology, Weill Cornell Medical College, New York, NY
| |
Collapse
|
43
|
Sethi TK, Reddy NM. Treatment of newly diagnosed primary central nervous system lymphoma: current and emerging therapies. Leuk Lymphoma 2018; 60:6-18. [DOI: 10.1080/10428194.2018.1466296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Tarsheen K. Sethi
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nishitha M. Reddy
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
44
|
Szczepanek D, Wąsik-Szczepanek E, Szymczyk A, Gromek T, Grywalska E, Podhorecka M, Hus M. Intraventricular treatment of secondary central nervous system lymphoma - Case study and literature overview. Neurol Neurochir Pol 2018; 52:410-414. [PMID: 29703403 DOI: 10.1016/j.pjnns.2018.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/12/2017] [Accepted: 01/27/2018] [Indexed: 11/20/2022]
Abstract
Secondary nervous system lymphoma (SCNSL) is a rare extranodal form of non-Hodgkin lymphoma (NHL). This applies to a particular form of lymphoma that does not originally derive from the central nervous system (CNS); it can be both an isolated form of relapse or a systemic part of disease progression. Due to poor prognosis and a lack of established algorithms of therapeutic procedures, it is a big challenge for physicians from many specializations. In our study, we present an interesting case of a patient with a relapsed form of SCNSL for whom a unique form of treatment was used - intraventricular administration of rituximab and methotrexate.
Collapse
Affiliation(s)
- Dariusz Szczepanek
- Chair and Department of Neurosurgery and Paediatric Neurosurgery Medical University of Lublin, Poland
| | - Ewa Wąsik-Szczepanek
- Chair and Department of Haematooncology and Bone Marrow Transplantation Medical University of Lublin, Poland
| | - Agnieszka Szymczyk
- Chair and Department of Haematooncology and Bone Marrow Transplantation Medical University of Lublin, Poland; Independent Clinical Transplantology Unit Medical University of Lublin, Poland.
| | - Tomasz Gromek
- Chair and Department of Haematooncology and Bone Marrow Transplantation Medical University of Lublin, Poland
| | - Ewelina Grywalska
- Chair and Department of Clinical Immunology Medical University of Lublin, Poland
| | - Monika Podhorecka
- Chair and Department of Haematooncology and Bone Marrow Transplantation Medical University of Lublin, Poland
| | - Marek Hus
- Chair and Department of Haematooncology and Bone Marrow Transplantation Medical University of Lublin, Poland
| |
Collapse
|
45
|
Sun X, Liu J, Wang Y, Bai X, Chen Y, Qian J, Zhu H, Liu F, Qiu X, Sun S, Ji N, Liu Y. Methotrexate-cytarabine-dexamethasone combination chemotherapy with or without rituximab in patients with primary central nervous system lymphoma. Oncotarget 2018; 8:49156-49164. [PMID: 28467782 PMCID: PMC5564757 DOI: 10.18632/oncotarget.17101] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/02/2017] [Indexed: 12/26/2022] Open
Abstract
Purpose High-dose methotrexate based chemotherapy is the standard treatment for patients with newly diagnosed primary central nervous system lymphoma (PCNSL). The role of rituximab is controversial because of its large size, which limits its penetration of the blood-brain barrier. In this study, we investigated the efficacy and tolerability of adding rituximab to methotrexate-cytarabine-dexamethasone combination therapy (RMAD regimen). Results The patients treated with RMAD had a complete remission rate of 66.7% after induction chemotherapy; this rate was only 33.3% in patients treated with MAD alone (p = .011). The most common grade 1–3 adverse events were similar and included hematologic toxicity, increased aminotransferase levels, and gastrointestinal reactions. Multivariate analysis revealed that rituximab treatment was associated with longer progression-free survival (PFS, p = .005) but not overall survival (OS). Additionally, we observed that elevated serum lactate dehydrogenase was associated with shorter OS and PFS. Materials and Methods We retrospectively analyzed 60 immunocompetent patients with newly diagnosed PCNSL at Beijing Tiantan Hospital, Capital Medical University from January 2010 to June 2016. Twenty-four patients received 3–6 courses of 3.5 g/m2 methotrexate on day 1; 0.5–1 g/m2 cytarabine on day 2; and 5–10 mg dexamethasone on days 1, 2 and 3. Thirty-six patients received the same combination plus rituximab 375 mg/m2 on day 0. All patients repeated the treatment every 3 weeks. Conclusions High-dose methotrexate based chemotherapy with rituximab yields a higher complete remission rate and does not increase serious toxicities. PFS benefits from the addition of rituximab. OS has an increasing trend in patients treated with rituximab without statistical significance.
Collapse
Affiliation(s)
- Xuefei Sun
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Liu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaming Wang
- Department of Neurosurgery, Navy General Hospital, Beijing, China
| | - Xueyan Bai
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuedan Chen
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Qian
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hong Zhu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fusheng Liu
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoguang Qiu
- Department of Radiation Therapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shengjun Sun
- Neuroimaging Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuanbo Liu
- Department of Hematology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Murthy H, Anasetti C, Ayala E. Diagnosis and Management of Leukemic and Lymphomatous Meningitis. Cancer Control 2018; 24:33-41. [DOI: 10.1177/107327481702400105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Hemant Murthy
- From the University of South Florida Morsani College of Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Claudio Anasetti
- From the University of South Florida Morsani College of Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Ernesto Ayala
- From the University of South Florida Morsani College of Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
47
|
Citterio G, Calimeri T, Ferreri AJM. Challenges and prospects in the diagnosis and treatment of primary central nervous system lymphoma. Expert Rev Neurother 2018; 18:379-393. [PMID: 29633883 DOI: 10.1080/14737175.2018.1462700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Primary central nervous system lymphoma (PCNSL) retains peculiar biological and clinical characteristics and a worse prognosis with respect to other comparable lymphomas. The need for high doses of chemotherapy to achieve valid drug concentrations in cerebral tissues and/or radiotherapy results in severe treatment-related toxicities, mainly neurologic, which are frequently as disabling as the disease itself.Areas covered: Several emerging combined therapies are addressed that focus on treating PCNSL. The prognosis has improved in the last years but several questions remain unanswered and the research of more effective therapies goes on. Information and data were obtained from direct authors' experience and a PubMed search of recent peer-reviewed original articles, review articles, and clinical guidelines.Expert commentary: The substantial progress observed in PCNSL has to be ascribed to a carefully combination of standard chemotherapeutic drugs. High-dose methotrexate-based polychemotherapy followed by mainteinance therapy offers one of the best chances to control the disease. Major issues that deserve many efforts by researchers are the definition of optimal consolidation treatment and a shared management of specific conditions such as elderly population and intra-ocular localization.
Collapse
Affiliation(s)
- Giovanni Citterio
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Teresa Calimeri
- Unit of Lymphoid Malignancies, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Andrés J M Ferreri
- Unit of Lymphoid Malignancies, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
48
|
Rubenstein JL. Biology of CNS lymphoma and the potential of novel agents. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:556-564. [PMID: 29222305 PMCID: PMC6053314 DOI: 10.1182/asheducation-2017.1.556] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Primary and secondary CNS lymphomas are aggressive brain tumors that pose an immense challenge to define in terms of molecular pathogenesis, as well as to effectively treat. During the past 10 years improvements in survival have been achieved with the implementation of anti-CD20 immunotherapy and optimization of dose-intensive consolidation strategies. The applications of whole-exome sequencing, comparative genomic hybridization, transcriptional profiling, and examination of the tumor microenvironment, particularly in the context of clinical investigation, provide insights that create a roadmap for the development and implementation of novel targeted agents for this disease. A body of genetic evidence strongly suggested that primary CNS lymphomas (PCNSLs) are likely largely dependent on NF-κB prosurvival signals, with enrichment of mutations involving the B-cell receptor pathway, in particular myeloid differentiation primary response 88 and cluster of differentiation 79B. The first set of early-phase investigations that target NF-κB in PCNSL have now been completed and support the NF-κB hypothesis but at the same time reveal that much work needs to be done to translate these results into meaningful advances in survival for a large fraction of patients. Insights into secondary prosurvival pathways that mediate drug resistance is a priority for investigation. Similarly, further evaluation of the immune-suppressive mechanisms in the CNS lymphoma tumor microenvironment is requisite for progress. Combinatorial interventions that promote the antitumor immune response have significant potential. With increasing availability of targeted agents, there is also a need to develop more sensitive imaging tools, not only to detect this highly invasive brain neoplasm but also potentially to define an evolving molecular phenotype to facilitate precision medicine.
Collapse
Affiliation(s)
- James L Rubenstein
- Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
49
|
Qian L, Tomuleasa C, Florian IA, Shen J, Florian IS, Zdrenghea M, Dima D. Advances in the treatment of newly diagnosed primary central nervous system lymphomas. Blood Res 2017; 52:159-166. [PMID: 29043230 PMCID: PMC5641507 DOI: 10.5045/br.2017.52.3.159] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/29/2017] [Indexed: 12/18/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a type of highly invasive non-Hodgkin lymphoma. With a growing number of organ transplantation and immunosuppressant therapy, the incidence of PCNSL has been growing rapidly in recent years, which is attributed to the increased incidence of HIV/AIDS, a prominent risk factor for developing PCNSL. The rising rate of PCNSL incidence is the highest among the intracranial tumors. In the past 20 years, dozens of clinical trials related to PCNSL have been registered, but adequate therapeutics are still challenging. Currently, the chemotherapy regimens based on high-dose methotrexate and whole-brain radiotherapy are the two main therapeutic options; however, the toxicity associated with those is the main problem that challenges medical researchers. Novel agents and therapeutic strategies have been developed in recent years. In the current review, we describe advances in the treatment of PCNSL and discuss novel therapeutic approaches currently in development, such as the use of rituximab, disruption of the blood-brain barrier, and state-of-the-art radiotherapy.
Collapse
Affiliation(s)
- Liren Qian
- Department of Hematology, Navy General Hospital of PLA, Beijing, China
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | | | - Jianliang Shen
- Department of Hematology, Navy General Hospital of PLA, Beijing, China
| | - Ioan-Stefan Florian
- Department of Neurosurgery, Emergency University Hospital, Cluj Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| |
Collapse
|
50
|
Song Y, Wen Y, Xue W, Zhang Y, Zhang M. Effect of rituximab on primary central nervous system lymphoma: a meta-analysis. Int J Hematol 2017; 106:612-621. [PMID: 28900847 DOI: 10.1007/s12185-017-2316-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 11/29/2022]
Abstract
The effect of rituximab on primary central nervous system lymphoma (PCNSL) is controversial. We performed this meta-analysis to assess the efficacy of treatment with or without rituximab for PCNSL. We first conducted a search for qualified studies using PubMed, the Cochrane Library, and the Web of Science. The meta-analysis was conducted to compare odds ratios (ORs) with the corresponding 95% confidence interval (95% CI) for complete remission (CR) rate, progression-free survival (PFS), and overall survival (OS) using Review Manager 5.0. We included two randomized clinical trials and six retrospective studies in this meta-analysis. The results of our statistical analysis show that the use of rituximab was closely correlated with a higher CR (OR 1.70, 95% CI 1.17-2.46, P = 0.005), 2-year PFS (OR 2.11, 95% CI 1.08-4.11, P = 0.03), 5-year PFS (OR 2.54, 95% CI 1.64-3.93, P < 0.0001), 2-year OS (OR 2.40, 95% CI 1.73-3.34, P < 0.00001), and 5-year OS (OR 2.87, 95% CI 2.02-4.08, P < 0.00001). These results may help to inform therapeutic strategies including the use of rituximab and to improve therapeutic planning for PCNSL patients.
Collapse
Affiliation(s)
- Yue Song
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Street (East), Erqi District, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yibo Wen
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.,Department of Urodynamics Centre, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Weili Xue
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Street (East), Erqi District, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanjie Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Street (East), Erqi District, Zhengzhou, 450000, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mingzhi Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Street (East), Erqi District, Zhengzhou, 450000, Henan, China. .,Lymphoma Diagnosis and Treatment Centre of Henan Province, Zhengzhou, 450000, Henan, China.
| |
Collapse
|