1
|
Nishida Y, Nonobe N, Kidokoro H, Kato T, Takeichi T, Ikuta K, Urakawa H, Sakai T, Koike H, Fujito T, Imagama S. Selumetinib for symptomatic, inoperable plexiform neurofibromas in pediatric patients with neurofibromatosis type 1: the first single-center real-world case series in Japan. Jpn J Clin Oncol 2025:hyae184. [PMID: 39756805 DOI: 10.1093/jjco/hyae184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/15/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE In Japan, selumetinib is used in pediatric patients with neurofibromatosis type 1 (NF1) and symptomatic inoperable plexiform neurofibroma (PN). However, there have been no real-world reports on Japanese patients. In this study, we reported a single-center, short-term experience with selumetinib after its approval in Japan. METHODS We prospectively collected data from 11 pediatric NF1 patients with symptomatic, inoperable PN who were initiated on selumetinib between November 2022 and May 2023; the selumetinib was administered by the same physician. Various patient factors, tumors, dose and efficacy of selumetinib, and adverse events (AE) were investigated. RESULTS Of 11 patients included, 7 were male, with a mean age of 14 years. The sites of symptomatic main PN included the head and neck, pelvis to lower extremities, and paraspinal lesions in five, three, and three patients, respectively. The median maximum diameter of the main PN was 91 mm, and the median follow-up duration was 19 months. All patients with pain or motor dysfunction experienced symptom improvement after treatment, and the tumors tended to shrink in 7 of the 11 patients (64%). Among the six patients with disfigurements, only one experienced improvement. Of 59 AEs observed, 58 (98%) were grades 1 and 2, and 5 patients (46%) underwent temporary selumetinib withdrawal due to AEs. One patient discontinued the drug (9%) because of rash dermatitis. CONCLUSIONS Despite the relatively short-term results, no serious AEs were observed, and many patients benefited from selumetinib treatment. In some patients, administration was discontinued or interrupted because of the balance between benefits and AEs, and further data are needed to better understand the general safety and efficacy of selumetinib.
Collapse
Affiliation(s)
- Yoshihiro Nishida
- Department of Rehabilitation Medicine, Nagoya University Hospital, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
- Rare Cancer Center, Nagoya University Hospital, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Norie Nonobe
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Taichi Kato
- Department of Pediatrics, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Takuya Takeichi
- Department of Dermatology, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Kunihiro Ikuta
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Hiroshi Urakawa
- Advanced Medicine, Nagoya University Hospital, 65-Tsrumai, Showa, Nagoya, 466-8560, Japan
| | - Tomohisa Sakai
- Rare Cancer Center, Nagoya University Hospital, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Hiroshi Koike
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Takeo Fujito
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65-Tsurumai, Showa, Nagoya, 466-8560, Japan
| |
Collapse
|
2
|
Zipfel J, Tellermann J, Ferraris KP, Grimm F, Bornemann A, Bender B, Dittmann H, Schäfer J, Nikolaou K, Ladurner R, Steger V, Tatagiba M, Schuhmann MU, Gugel I. Perioperative Observations and Outcome in Surgical Treatment of Malignant Peripheral Nerve Sheath Tumors. Cancers (Basel) 2024; 16:3757. [PMID: 39594712 PMCID: PMC11592335 DOI: 10.3390/cancers16223757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES This retrospective observational study aimed to investigate the perioperative outcome in Malignant Peripheral Nerve Sheath Tumors (MPNSTs) with and without relation to Neurofibromatosis Type 1 (NF1) and to detect possible influencing factors. METHODS Clinical reports, histopathological evaluations, imaging, and treatment characteristics were reviewed in 35 operated MPNSTs in 33 patients. Possible predictive valuables included disease type, preoperative tumor volume, SUV and MIB-1 proliferation index, resection margins, the presence of metastasis, and whether radio-/chemotherapy was received. RESULTS Patients with NF1 were younger (mean age: 29 ± 13, 8-54 years) than sporadic cases (mean age: 45 ± 13, 24-67 years) and exhibited significantly larger preoperative tumor volumes (mean 299 vs. 18 cm3, p = 0.048). Most tumors were located in the facial/cervical/neck area (34%, n = 12), followed by the trunk (31%, n = 11), lower extremity (17%, n = 6), upper extremity (14%, n = 5), and intraspinal area (3%, n = 1). NF1-associated MPNSTs appeared predominantly on the trunk (39%) and sporadically in the facial/cervical/neck area (50%). Complete resection was possible in 66% and an improvement in or stability of function was achieved in most cases (motor 69%, sensory 74%), as well as a decrease in pain intensity (63%). NF1-associated MPNSTs exhibited more severe pain scores (median VRS scale 2, p = 0.002) compared to sporadic tumors (median VRS scale 0.5). Sporadic MPNSTs located at the head/facial/brachial plexus and upper extremities exhibited better preoperative functions compared to those on the lower extremities. In 12 cases with available [18F]FDG PET, the mean preoperative SUV (9.8 ± 7.2) positively correlated with the mean maximum MIB-1 index (34 ± 26%, p = 0.005) and the mean preoperative tumor volume (474.7 ± 68.6 cm3, p = 0.047). The overall survival (OS) was significantly longer in tumors with higher resection extents (R0, p = 0.01) and without accompanying metastasis (p = 0.046), and tended to be longer, but not significantly so, in sporadic MPNSTs. In six and seven tumors, with R1/R2 resection margins and present metastasis, respectively, solid or combined neo-/adjuvant radio-/chemotherapy led to a significantly shorter OS (p = 0.014). CONCLUSIONS NF1-associated MPNSTs have larger tumor volumes, higher SUVs and MIB-1 proliferation indices, and a shorter overall survival period. Nevertheless, surgery can improve symptoms, particularly medication-resistant pain, and should also be considered in advanced disease for symptom control/improvement.
Collapse
Affiliation(s)
- Julian Zipfel
- Department of Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Centre of Neurofibromatosis and Rare Diseases, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Section of Pediatric Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Division of Pediatric Neurosurgery, BC Children’s Hospital, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Jonas Tellermann
- Department of Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Section of Pediatric Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Kevin Paul Ferraris
- Division of Pediatric Neurosurgery, BC Children’s Hospital, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Florian Grimm
- Department of Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Antje Bornemann
- Department of Pathology and Neuropathology and Comprehensive Cancer Center, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Benjamin Bender
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Helmut Dittmann
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Jürgen Schäfer
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Ruth Ladurner
- Department of General, Visceral and Transplantation Surgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, BW 72076 Tübingen, Germany
| | - Volker Steger
- Department of Thoracic and Cardiovascular Surgery, German Cardiac Competence Center, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Marcos Tatagiba
- Department of Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Centre of Neurofibromatosis and Rare Diseases, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Martin U. Schuhmann
- Department of Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Centre of Neurofibromatosis and Rare Diseases, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Section of Pediatric Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
| | - Isabel Gugel
- Department of Neurosurgery, University Hospital Tübingen, BW 72076 Tübingen, Germany
- Centre of Neurofibromatosis and Rare Diseases, University Hospital Tübingen, BW 72076 Tübingen, Germany
| |
Collapse
|
3
|
Lemberg KM, Gross AM, Sproule LM, Liewehr DJ, Dombi E, Baldwin A, Steinberg SM, Bornhorst M, Lodish M, Blakeley JO, Widemann BC. Anthropometric measurements of children with neurofibromatosis type I: impact of plexiform neurofibroma volume and treatment. Pediatr Res 2024:10.1038/s41390-024-03474-z. [PMID: 39198589 DOI: 10.1038/s41390-024-03474-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND In children and adolescents/young adults (CAYA) with neurofibromatosis type I (NF1), associations between anthropometric measurements, plexiform neurofibroma (pNF) tumor volume (TV), and treatment history are unknown. METHODS We retrospectively investigated anthropometrics in CAYA on the National Cancer Institute (NCI) NF1 Natural History Study who had pNF TV assessed by imaging (n = 106). We determined CDC height/weight percentiles and estimated Preece-Baines (PB) height growth curve parameters. We evaluated variables that could impact height/weight including: (1) pNF volume, (2) pNF directed therapy, and (3) serum IGF-1. RESULTS 23% of males and 20% of females had height <5th percentile; 13% of males had weight <5th percentile. Estimated median final adult height for males was 171.6 cm (CDC 23rd percentile) and for females was 156.2 cm (CDC 14th percentile). Inverse associations between height and weight percentiles and pNF volume were observed (Spearman's r = -0.277, -0.216, respectively). Estimated median final height was not meaningfully affected by patients who received pNF-directed treatment with MEK inhibitor. 52% of low serum IGF-1 measurements were concurrent with a height percentile <5th. CONCLUSIONS Greater than expected percentages of patients had height/weight <5th percentile, and median final adult heights were IMPACT STATEMENT Children and adolescents/young adults with neurofibromatosis type I (NF1) seen at a research hospital have lower height and weight percentiles than normative populations. Growth percentiles are inversely associated with plexiform neurofibroma tumor volumes and impacted little by MEKi treatment history in this subset of patients. These findings align with prior investigations of growth in the NF1 population but are the first to examine the association with tumor burden.
Collapse
Affiliation(s)
- Kathryn M Lemberg
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Pediatric Oncology Branch, Center for Cancer Research, Bethesda, MD, USA.
| | - Andrea M Gross
- Pediatric Oncology Branch, Center for Cancer Research, Bethesda, MD, USA
| | | | - David J Liewehr
- Biostatistics and Data Management Section, Center for Cancer Research, Bethesda, MD, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, Bethesda, MD, USA
| | - Andrea Baldwin
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, Bethesda, MD, USA
| | - Miriam Bornhorst
- Gilbert Family Neurofibromatosis Institute, Children's National Hospital, Washington, DC, USA
- Center for Cancer & Blood Disorders, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Maya Lodish
- Division of Pediatric Endocrinology, University of California San Francisco, San Francisco, CA, USA
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
4
|
Bashiri FA, Hundallah K, Abukhaled M, Alyahya MM, Al Futaisi A, Alshowaeir D, Al Tawari A, Abdullah S, Maaz AUR, AlShamsi ET, Alshuaibi W, Alotaibi F, Aldhalaan H. Diagnosis and management of neurofibromatosis type 1 in Arabian Gulf Cooperation Council Region: challenges and recommendations. Front Oncol 2024; 14:1323176. [PMID: 39257551 PMCID: PMC11385870 DOI: 10.3389/fonc.2024.1323176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is a complex multisystem genetic disorder that requires long-term, age-specific monitoring and multidisciplinary care. NF1 symptom burden can significantly affect the quality of life and impose a substantial economic burden on patients and their families. The approval and widespread availability of mitogen-activated protein kinase (MEK) inhibitors such as selumetinib for NF1-related plexiform neurofibromas have revolutionized the standard of care for patients with NF1, however their effective utilization hinges on early recognition of NF1. We present a consensus manuscript describing the challenges observed in the Arabian Gulf Cooperation Council (GCC) for diagnosing and managing NF1. Experts from the GCC also present recommendations for the early recognition and management of NF1 and its complications. A referral pathway that can play a crucial role in helping primary healthcare providers refer their patients to experts is also proposed. Increasing the availability and accessibility of genetic testing at an affordable cost and optimizing personalized NF1 care are essential for NF1 management. Developing regional guidelines for NF1 management and establishing NF1 centers of excellence may facilitate better care and outcomes for patients with NF1 in the GCC region.
Collapse
Affiliation(s)
- Fahad A. Bashiri
- Division of Pediatric Neurology, Department of Pediatrics, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | | | - Musaad Abukhaled
- Neuroscience Centre, King Faisal Specialist Hospital and Research Centre, College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| | - Mossaed Mohammed Alyahya
- Consultant of Neuro-oncology and Neuromuscular-neurology, Department of Oncology, King Faisal Specialist Hospital and Research Centre, Department of Neuroscience, King Faisal Specialist Hospital and Research Centre, Al Faisal University, Riyadh, Saudi Arabia
| | - Amna Al Futaisi
- Sultan Qaboos University, College of Medicine and Health Sciences, Muscat, Oman
| | - Daniah Alshowaeir
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Asmaa Al Tawari
- Pediatric Neurology Unit, Pediatric Department, AlSabah Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Shaker Abdullah
- Department of Oncology, King Faisal Specialist Hospital & Research Center – Jeddah (KFSHRC-Jed), Jeddah, Saudi Arabia
| | - Ata Ur Rehman Maaz
- HemOnc Division, Department of Child Health, Sidra Medicine, Doha, Qatar
| | - Eman Taryam AlShamsi
- Pediatric Hematology-Oncology Department, Al Jalila children’s specialty Hospital, Dubai, United Arab Emirates
| | - Walaa Alshuaibi
- Division of Medical Genetics, Department of Pediatrics, King Khalid University Hospital, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Alotaibi
- Neuroscience Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hesham Aldhalaan
- Department of Neurosciences, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Tamura R, Yamanobe Y, Fujioka M, Morimoto Y, Fukumura M, Nakaya M, Oishi Y, Sato M, Ueda R, Fujiwara H, Hikichi T, Noji S, Oishi N, Ozawa H, Ogawa K, Kawakami Y, Ohira T, Yoshida K, Toda M. Phase I/II Study of a Vascular Endothelial Growth Factor Receptor Vaccine in Patients With NF2-Related Schwannomatosis. J Clin Oncol 2024; 42:2578-2587. [PMID: 38776485 DOI: 10.1200/jco.23.02376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/15/2024] [Accepted: 03/21/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE The humanized antivascular endothelial growth factor (VEGF) antibody bevacizumab (Bev) is efficacious for the treatment of NF2-related schwannomatosis (NF2), previously known as neurofibromatosis type 2. This study evaluated the safety and efficacy of a VEGF receptor (VEGFR) vaccine containing VEGFR1 and VEGFR2 peptides in patients with NF2 with progressive schwannomas (jRCTs031180184). MATERIALS AND METHODS VEGFR1 and VEGFR2 peptides were injected subcutaneously into infra-axillary and inguinal regions, once a week for 4 weeks and then once a month for 4 months. The primary end point was safety. Secondary end points included tolerability, hearing response, imaging response, and immunologic response. RESULTS Sixteen patients with NF2 with progressive schwannomas completed treatment and were assessed. No severe vaccine-related adverse events occurred. Among the 13 patients with assessable hearing, word recognition score improved in five patients at 6 months and two at 12 months. Progression of average hearing level of pure tone was 0.168 dB/mo during the year of treatment period, whereas long-term progression was 0.364 dB/mo. Among all 16 patients, a partial response was observed in more than one schwannoma in four (including one in which Bev had not been effective), minor response in 5, and stable disease in 4. Both VEGFR1-specific and VEGFR2-specific cytotoxic T lymphocytes (CTLs) were induced in 11 patients. Two years after vaccination, a radiologic response was achieved in nine of 20 assessable schwannomas. CONCLUSION This study demonstrated the safety and preliminary efficacy of VEGFR peptide vaccination in patients with NF2. Memory-induced CTLs after VEGFR vaccination may persistently suppress tumor progression.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiharu Yamanobe
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
- Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
- Department of Molecular Genetics, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Mariko Fukumura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Nakaya
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yumiko Oishi
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Mizuto Sato
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Ueda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Hirokazu Fujiwara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | | | - Shinobu Noji
- Division of Cellular Signaling Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Oishi
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Department of Immunology, International University of Health and Welfare School of Medicine, Narita, Japan
| | - Takayuki Ohira
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Plotkin SR, Yohay KH, Nghiemphu PL, Dinh CT, Babovic-Vuksanovic D, Merker VL, Bakker A, Fell G, Trippa L, Blakeley JO. Brigatinib in NF2-Related Schwannomatosis with Progressive Tumors. N Engl J Med 2024; 390:2284-2294. [PMID: 38904277 DOI: 10.1056/nejmoa2400985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
BACKGROUND NF2-related schwannomatosis (NF2-SWN, formerly called neurofibromatosis type 2) is a tumor predisposition syndrome that is manifested by multiple vestibular schwannomas, nonvestibular schwannomas, meningiomas, and ependymomas. The condition is relentlessly progressive with no approved therapies. On the basis of preclinical activity of brigatinib (an inhibitor of multiple tyrosine kinases) in NF2-driven nonvestibular schwannoma and meningioma, data were needed on the use of brigatinib in patients with multiple types of progressive NF2-SWN tumors. METHODS In this phase 2 platform trial with a basket design, patients who were 12 years of age or older with NF2-SWN and progressive tumors were treated with oral brigatinib at a dose of 180 mg daily. A central review committee evaluated one target tumor and up to five nontarget tumors in each patient. The primary outcome was radiographic response in target tumors. Key secondary outcomes were safety, response rate in all tumors, hearing response, and patient-reported outcomes. RESULTS A total of 40 patients (median age, 26 years) with progressive target tumors (10 vestibular schwannomas, 8 nonvestibular schwannomas, 20 meningiomas, and 2 ependymomas) received treatment with brigatinib. After a median follow-up of 10.4 months, the percentage of tumors with a radiographic response was 10% (95% confidence interval [CI], 3 to 24) for target tumors and 23% (95% CI, 16 to 30) for all tumors; meningiomas and nonvestibular schwannomas had the greatest benefit. Annualized growth rates decreased for all tumor types during treatment. Hearing improvement occurred in 35% (95% CI, 20 to 53) of eligible ears. Exploratory analyses suggested a decrease in self-reported pain severity during treatment (-0.013 units per month; 95% CI, -0.002 to -0.029) on a scale from 0 (no pain) to 3 (severe pain). No grade 4 or 5 treatment-related adverse events were reported. CONCLUSIONS Brigatinib treatment resulted in radiographic responses in multiple tumor types and clinical benefit in a heavily pretreated cohort of patients with NF2-SWN. (Funded by the Children's Tumor Foundation and others; INTUITT-NF2 ClinicalTrials.gov number, NCT04374305.).
Collapse
Affiliation(s)
- Scott R Plotkin
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Kaleb H Yohay
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Phioanh L Nghiemphu
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Christine T Dinh
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Dusica Babovic-Vuksanovic
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Vanessa L Merker
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Annette Bakker
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Geoffrey Fell
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Lorenzo Trippa
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| | - Jaishri O Blakeley
- From Massachusetts General Hospital and Harvard Medical School (S.R.P., V.L.M.) and the Dana-Farber Cancer Institute (G.F., L.T.) - all in Boston; the NYU Grossman School of Medicine (K.H.Y.) and the Children's Tumor Foundation (A.B.) - both in New York; the University of California, Los Angeles, Los Angeles (P.L.N.); the University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami (C.T.D.); the Mayo Clinic, Rochester, MN (D.B.-V.); and Johns Hopkins University, Baltimore (J.O.B.)
| |
Collapse
|
7
|
Perrino MR, Ahmari N, Hall A, Jackson M, Na Y, Pundavela J, Szabo S, Woodruff TM, Dombi E, Kim MO, Köhl J, Wu J, Ratner N. C5aR plus MEK inhibition durably targets the tumor milieu and reveals tumor cell phagocytosis. Life Sci Alliance 2024; 7:e202302229. [PMID: 38458648 PMCID: PMC10923703 DOI: 10.26508/lsa.202302229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 03/10/2024] Open
Abstract
Plexiform neurofibromas (PNFs) are nerve tumors caused by loss of NF1 and dysregulation of RAS-MAPK signaling in Schwann cells. Most PNFs shrink in response to MEK inhibition, but targets with increased and durable effects are needed. We identified the anaphylatoxin C5a as increased in PNFs and expressed largely by PNF m acrophages. We defined pharmacokinetic and immunomodulatory properties of a C5aR1/2 antagonist and tested if peptide antagonists augment the effects of MEK inhibition. MEK inhibition recruited C5AR1 to the macrophage surface; short-term inhibition of C5aR elevated macrophage apoptosis and Schwann cell death, without affecting MEK-induced tumor shrinkage. PNF macrophages lacking C5aR1 increased the engulfment of dying Schwann cells, allowing their visualization. Halting combination therapy resulted in altered T-cell distribution, elevated Iba1+ and CD169+ immunoreactivity, and profoundly altered cytokine expression, but not sustained trumor shrinkage. Thus, C5aRA inhibition independently induces macrophage cell death and causes sustained and durable effects on the PNF microenvironment.
Collapse
Affiliation(s)
- Melissa R Perrino
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Niousha Ahmari
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Hall
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mark Jackson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Youjin Na
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jay Pundavela
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sara Szabo
- Departmentd of Pediatrics and Pediatric Pathology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, Australia
| | - Eva Dombi
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Mi-Ok Kim
- Department Biostatistics, University of California, San Francisco, CA, USA
| | - Jörg Köhl
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Institute for Systemic Inflammation Research, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
8
|
Shrivastava M, Emmanouil B, Mathew R, Halliday D, Parry A, Halliday J, Mackeith S. Stereotactic Radiosurgery and Radiotherapy for Vestibular Schwannoma in NF2-Related Schwannomatosis. Laryngoscope 2024; 134:2364-2371. [PMID: 37983868 DOI: 10.1002/lary.31180] [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: 08/07/2023] [Revised: 10/11/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVES To determine the long-term control rates and hearing outcomes for growing vestibular schwannoma in NF2-related schwannomatosis (NF2) treated with stereotactic radiosurgery (SRS) and fractionated radiotherapy (FRT). METHODS Retrospective review of all patients treated with SRS/FRT between 1986 and2021 from a tertiary NF2 unit. Overall tumor control was defined as: (1) growth control (growth failure was defined as growth in any dimension of 3 millimetres or more from baseline post-SRS/FRT), and (2) treatment control (no need for further intervention). Loss of serviceable hearing was defined as a drop in speech discrimination score below 50% after SRS/FRT. RESULTS There were 81 cases, with a mean duration of follow-up of 125 months. Overall control rate was 72% (58/81), with 80% (65/81) growth control and 74% (60/81) treatment control. There was a 5-year actuarial survival of 77% and 10-year survival of 71%. Forty-three percent (30/69) of cases did not have serviceable hearing at baseline. Of those remaining, 49% (19/39) preserved serviceable hearing during follow-up at a mean of 106 months. Actuarial survival for preservation of serviceable hearing at 5 and 10 years was 69% and 53%. There were poorer outcomes with increasing genetic severity, and with baseline tumor size >3 cm. No cases of SRS/FRT-related malignancy were identified at a mean follow-up of 10 years. CONCLUSION Stereotactic radiosurgery/fractionated radiotherapy are an effective option to treat growing vestibular schwannoma in patients with NF2 with the potential for hearing preservation in a proportion of patients. LEVEL OF EVIDENCE 4-Case Series Laryngoscope, 134:2364-2371, 2024.
Collapse
Affiliation(s)
- Manu Shrivastava
- Department of Ear, Nose and Throat Surgery, Oxford University Hospitals, Oxford, UK
| | - Beatrice Emmanouil
- NHS England, Wellington House, London, UK
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rajeev Mathew
- Department of Ear, Nose and Throat Surgery, Oxford University Hospitals, Oxford, UK
| | | | - Allyson Parry
- Department of Neurology, Oxford University Hospitals, Oxford, UK
| | - Jane Halliday
- Department of Neurosurgery, Oxford University Hospitals, Oxford, UK
| | - Samuel Mackeith
- Department of Ear, Nose and Throat Surgery, Oxford University Hospitals, Oxford, UK
| |
Collapse
|
9
|
Douwes JPJ, Hensen EF, Jansen JC, Gelderblom H, Schopman JE. Bevacizumab Treatment for Patients with NF2-Related Schwannomatosis: A Single Center Experience. Cancers (Basel) 2024; 16:1479. [PMID: 38672561 PMCID: PMC11047890 DOI: 10.3390/cancers16081479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
(1) Background: NF2-related schwannomatosis, characterized by the development of bilateral vestibular schwannomas, often necessitates varied treatment approaches. Bevacizumab, though widely utilized, demonstrates variable effectiveness on hearing and tumor growth. At the same time, (serious) adverse events have been frequently reported. (2) Methods: A single center retrospective study was conducted, on NF2-related schwannomatosis patients treated with bevacizumab from 2013 to 2023, with the aim to assess treatment-related and clinical outcomes. Outcomes of interest comprised hearing, radiologic response, symptoms, and adverse events. (3) Results: Seventeen patients received 7.5 mg/kg bevacizumab for 7.1 months. Following treatment, 40% of the patients experienced hearing improvement, 53%, stable hearing, and 7%, hearing loss. Vestibular schwannoma regression occurred in 31%, and 69% remained stable. Further symptomatic improvement was reported by 41%, stable symptoms by 47%, and worsened symptoms by 12%. Treatment discontinuation due to adverse events was observed in 29% of cases. Hypertension (82%) and fatigue (29%) were most frequently reported, with no occurrences of grade 4/5 toxicities. (4) Conclusion: Supporting previous studies, bevacizumab demonstrated positive effects on hearing, tumor control, and symptoms in NF2-related schwannomatosis, albeit with common adverse events. Therefore, careful consideration of an appropriate management strategy is warranted.
Collapse
Affiliation(s)
- Jules P. J. Douwes
- Department of Otorhinolaryngology–Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (E.F.H.); (J.C.J.)
| | - Erik F. Hensen
- Department of Otorhinolaryngology–Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (E.F.H.); (J.C.J.)
| | - Jeroen C. Jansen
- Department of Otorhinolaryngology–Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (E.F.H.); (J.C.J.)
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (H.G.); (J.E.S.)
| | - Josefine E. Schopman
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (H.G.); (J.E.S.)
| |
Collapse
|
10
|
Nghiemphu PL, Vitte J, Dombi E, Nguyen T, Wagle N, Ishiyama A, Sepahdari AR, Cachia D, Widemann BC, Brackmann DE, Doherty JK, Kalamarides M, Giovannini M. Imaging as an early biomarker to predict sensitivity to everolimus for progressive NF2-related vestibular schwannoma. J Neurooncol 2024; 167:339-348. [PMID: 38372904 PMCID: PMC11023969 DOI: 10.1007/s11060-024-04596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/01/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE NF2-related schwannomatosis (NF2) is characterized by bilateral vestibular schwannomas (VS) often causing hearing and neurologic deficits, with currently no FDA-approved drug treatment. Pre-clinical studies highlighted the potential of mTORC1 inhibition in delaying schwannoma progression. We conducted a prospective open-label, phase II study of everolimus for progressive VS in NF2 patients and investigated imaging as a potential biomarker predicting effects on growth trajectory. METHODS The trial enrolled 12 NF2 patients with progressive VS. Participants received oral everolimus daily for 52 weeks. Brain imaging was obtained quarterly. As primary endpoint, radiographic response (RR) was defined as ≥ 20% decrease in target VS volume. Secondary endpoints included other tumors RR, hearing outcomes, drug safety and quality of life (QOL). RESULTS Eight participants completed the trial and four discontinued the drug early due to significant volumetric VS progression. After 52 weeks of treatment, the median annual VS growth rate decreased from 77.2% at baseline to 29.4%. There was no VS RR and 3 of 8 (37.5%) participants had stable disease. Decreased or unchanged VS volume after 3 months of treatment was predictive of stabilization at 12 months. Seven of eight participants had stable hearing during treatment except one with a decline in word recognition score. Ten of twelve participants reported only minimal changes to their QOL scores. CONCLUSIONS Volumetric imaging at 3 months can serve as an early biomarker to predict long-term sensitivity to everolimus treatment. Everolimus may represent a safe treatment option to decrease the growth of NF2-related VS in patients who have stable hearing and neurological condition. TRN: NCT01345136 (April 29, 2011).
Collapse
Affiliation(s)
- Phioanh Leia Nghiemphu
- Department of Neurology, UCLA Neuro‑Oncology Program, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, 675 Charles E Young Dr. S, MRL 2240, Los Angeles, CA, 90095-7286, USA
| | - Eva Dombi
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Thien Nguyen
- Department of Neurology, UCLA Neuro‑Oncology Program, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Division of Pediatric Hematology, Oncology, Stem Cell Transplant and Regenerative Medicine, Stanford University, Palo Alto, CA, USA
| | - Naveed Wagle
- Department of Medicine, Division of Medical Oncology, Norris Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Translational Neurosciences, Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Akira Ishiyama
- Department of Head and Neck Surgery, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, 675 Charles E Young Dr. S, MRL 2240, Los Angeles, CA, 90095-7286, USA
| | - Ali R Sepahdari
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Diagnostic Neuroradiology, Scripps Clinic Medical Group, La Jolla, CA, USA
| | - David Cachia
- Department of Neurosurgery, Division of Neuro-oncology, Medical University of South Carolina, Charleston, SC, USA
- Department of Medicine, Division of Hematology/Oncology, University of Massachusetts, Worcester, MA, USA
| | | | - Derald E Brackmann
- Department of Otolaryngology and Neurotology, House Clinic and Research Institute, Los Angeles, CA, USA
| | - Joni K Doherty
- Center for Neural Tumor Research, House Research Institute, Los Angeles, CA, USA
- Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, CA, USA
| | - Michel Kalamarides
- Department of Neurosurgery, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine and Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles, 675 Charles E Young Dr. S, MRL 2240, Los Angeles, CA, 90095-7286, USA.
| |
Collapse
|
11
|
Kiaei DS, El-Jalbout R, Décarie JC, Perreault S, Dehaes M. Development of a semi-automatic segmentation technique based on mean magnetic resonance imaging intensity thresholding for volumetric quantification of plexiform neurofibromas. Heliyon 2024; 10:e23445. [PMID: 38173515 PMCID: PMC10761559 DOI: 10.1016/j.heliyon.2023.e23445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Rationale and objectives Plexiform neurofibromas (PNs) are peripheral nerve tumors that occur in 25-50 % of patients with neurofibromatosis type 1. PNs may have complex, diffused, and irregular shapes. The objective of this work was to develop a volumetric quantification method for PNs as clinical assessment is currently based on unidimensional measurement. Materials and methods A semi-automatic segmentation technique based on mean magnetic resonance imaging (MRI) intensity thresholding (SSTMean) was developed and compared to a similar and previously published technique based on minimum image intensity thresholding (SSTMini). The performance (volume and computation time) of the two techniques was compared to manual tracings of 15 tumors of different locations, shapes, and sizes. Performance was also assessed using different MRI sequences. Reproducibility was assessed by inter-observer analysis. Results When compared to manual tracing, quantification performed with SSTMean was not significantly different (mean difference: 1.2 %), while volumes computed by SSTMini were significantly different (p < .0001, mean difference: 13.4 %). Volumes quantified by SSTMean were also significantly different than the ones assessed by SSTMini (p < .0001). Using SSTMean, volumes quantified with short TI inversion recovery, T1-, and T2-weighted imaging were not significantly different. Computation times used by SSTMean and SSTMini were significantly lower than for manual segmentation (p < .0001). The highest difference measured by two users was 8 cm3. Conclusion Our method showed accuracy compared to a current gold standard (manual tracing) and reproducibility between users. The refined segmentation threshold and the possibility to define multiple regions-of-interest to initiate segmentation may have contributed to its performance. The versatility and speed of our method may prove useful to better monitor volumetric changes in lesions of patients enrolled in clinical trials to assessing response to therapy.
Collapse
Affiliation(s)
- Dorsa Sadat Kiaei
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada
- Research Center, CHU Sainte-Justine Hospital University Centre, Montréal, Canada
| | - Ramy El-Jalbout
- Research Center, CHU Sainte-Justine Hospital University Centre, Montréal, Canada
- Department of Radiology, Radio-oncology and Nuclear Medicine, University of Montreal, Montreal, Canada
| | - Jean-Claude Décarie
- Department of Radiology, Radio-oncology and Nuclear Medicine, University of Montreal, Montreal, Canada
| | - Sébastien Perreault
- Research Center, CHU Sainte-Justine Hospital University Centre, Montréal, Canada
- Department of Neurosciences, University of Montreal, Montreal, Canada
| | - Mathieu Dehaes
- Institute of Biomedical Engineering, University of Montréal, Montréal, Canada
- Research Center, CHU Sainte-Justine Hospital University Centre, Montréal, Canada
- Department of Radiology, Radio-oncology and Nuclear Medicine, University of Montreal, Montreal, Canada
| |
Collapse
|
12
|
Passos J, Soares MP, Salgado D, Nunes S, Cavaco D, Garrido PM, Coutinho M, Carvalho IP, Vilares M, Ferreira M, Lacerda C. A single-center case study series assessing the effect of selumetinib use in patients with neurofibromatosis-related plexiform neurofibromas. Neurooncol Adv 2024; 6:vdae177. [PMID: 39605314 PMCID: PMC11600336 DOI: 10.1093/noajnl/vdae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is a common genetic disorder of phenotypic variability with age-dependent penetrance. This study describes the diagnosis, clinical characterization, management, and outcomes of a large patient cohort with plexiform neurofibroma (PN) treated with selumetinib in a real-world clinical setting. Methods This single-center observational study consecutively enrolled patients with NF1-PN treated with selumetinib from April 2018 to 2023. Data on clinical features, tumor types and locations, and results from genetic tests were recorded at baseline; details of disease management with selumetinib and surgical intervention and disease evolution including imaging data and evaluations of pain and function were documented. Results Overall, 54 patients with a median age (range) of 16.4 (4.5-58.0) years were enrolled. Most had cutaneous manifestations (88.9%), including cutaneous neurofibromas and PN. Patients underwent [18F]fluorodeoxyglucose (FDG)-PET/CT imaging before treatment to rule out malignant lesions. Initial evaluations included directed magnetic resonance imaging (MRI), which facilitated future comparisons and allowed for the assessment of PN resectability. Pharmacological treatment with selumetinib (with surgery, without surgery) resulted in the following proportion of patients achieving stable disease (58.8%, 54.3%), partial response (29.4%, 28.6%), and improved pain (58.8%, 37.1%), deformity (17.6%, 20.0%), and functional (17.6%, 20.0%) outcomes, respectively. Conclusions Results from this study demonstrate that NF1-PN can be managed effectively with selumetinib with surgical intervention in some patients. Most patients achieved tumor stability and improved symptom control, and the majority of patients continue under treatment. Effective diagnosis and management were achieved through individualized utility of FDG-PET/CT and MRI imaging and targeted resource allocation.
Collapse
Affiliation(s)
- João Passos
- Department of Neurology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Marta P Soares
- Department of Genetic Medicine, Centro Hospitalar Universitário Lisboa Norte, E.P.E., Lisbon, Portugal
| | - Duarte Salgado
- Department of Neurology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
- Unit of Pediatric Neuro-Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Sofia Nunes
- Unit of Pediatric Neuro-Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Daniela Cavaco
- Department of Endocrinology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Pedro M Garrido
- Department of Dermatology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Mónica Coutinho
- Department of Radiology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Inês Patrocínio Carvalho
- Department of Nuclear Medicine, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Miguel Vilares
- Department of Head and Neck Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Mafalda Ferreira
- Department of Neurology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Cristina Lacerda
- Department of Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| |
Collapse
|
13
|
Telleman JA, Sneag DB, Visser LH. The role of imaging in focal neuropathies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 201:19-42. [PMID: 38697740 DOI: 10.1016/b978-0-323-90108-6.00001-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Electrodiagnostic testing (EDX) has been the diagnostic tool of choice in peripheral nerve disease for many years, but in recent years, peripheral nerve imaging has been used ever more frequently in daily clinical practice. Nerve ultrasound and magnetic resonance (MR) neurography are able to visualize nerve structures reliably. These techniques can aid in localizing nerve pathology and can reveal significant anatomical abnormalities underlying nerve pathology that may have been otherwise undetected by EDX. As such, nerve ultrasound and MR neurography can significantly improve diagnostic accuracy and can have a significant effect on treatment strategy. In this chapter, the basic principles and recent developments of these techniques will be discussed, as well as their potential application in several types of peripheral nerve disease, such as carpal tunnel syndrome (CTS), ulnar neuropathy at the elbow (UNE), radial neuropathy, brachial and lumbosacral plexopathy, neuralgic amyotrophy (NA), fibular, tibial, sciatic, femoral neuropathy, meralgia paresthetica, peripheral nerve trauma, tumors, and inflammatory neuropathies.
Collapse
Affiliation(s)
- Johan A Telleman
- Department of Neurology and Clinical Neurophysiology, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Darryl B Sneag
- Department of Radiology and Imaging, Hospital for Special Surgery, New York, NY, United States
| | - Leo H Visser
- Department of Neurology and Clinical Neurophysiology, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands.
| |
Collapse
|
14
|
Gross AM, Dombi E, Wolters PL, Baldwin A, Dufek A, Herrera K, Martin S, Derdak J, Heisey KS, Whitcomb PM, Steinberg SM, Venzon DJ, Fisher MJ, Kim A, Bornhorst M, Weiss BD, Blakeley JO, Smith MA, Widemann BC. Long-term safety and efficacy of selumetinib in children with neurofibromatosis type 1 on a phase 1/2 trial for inoperable plexiform neurofibromas. Neuro Oncol 2023; 25:1883-1894. [PMID: 37115514 PMCID: PMC10547508 DOI: 10.1093/neuonc/noad086] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Selumetinib shrank inoperable symptomatic plexiform neurofibromas (PN) in children with neurofibromatosis type 1 (NF1) and provided clinical benefit for many in our previously published phase 1/2 clinical trials (SPRINT, NCT01362803). At the data cutoff (DCO) of the prior publications, 65% of participants were still receiving treatment. This report presents up to 5 years of additional safety and efficacy data from these studies. METHODS This manuscript includes data from the phase 1 and phase 2, stratum 1 study which included participants with clinically significant PN-related morbidity. Participants received continuous selumetinib dosing (1 cycle = 28 days). Safety and efficacy data through February 27, 2021 are included. PN response assessed by volumetric magnetic resonance imaging analysis: Confirmed partial response (cPR) ≥20% decrease from baseline on 2 consecutive evaluations. Phase 2 participants completed patient-reported outcome measures assessing tumor pain intensity (Numeric Rating Scale-11) and interference of pain in daily life (pain interference index). RESULTS For the 74 children (median age 10.3 years; range 3-18.5) enrolled, overall cPR rate was 70% (52/74); median duration of treatment was 57.5 cycles (range 1-100). Responses were generally sustained with 59% (44) lasting ≥ 12 cycles. Tumor pain intensity (n = 19, P = .015) and pain interference (n = 18, P = .0059) showed durable improvement from baseline to 48 cycles. No new safety signals were identified; however, some developed known selumetinib-related adverse events (AEs) for the first time after several years of treatment. CONCLUSIONS With up to 5 years of additional selumetinib treatment, most children with NF1-related PN had durable tumor shrinkage and sustained improvement in pain beyond that previously reported at 1 year. No new safety signals were identified; however, ongoing monitoring for known selumetinib-related AEs is needed while treatment continues.
Collapse
Affiliation(s)
- Andrea M Gross
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Andrea Baldwin
- Leidos, Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Anne Dufek
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kailey Herrera
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Staci Martin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Joanne Derdak
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kara S Heisey
- Leidos, Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Patricia M Whitcomb
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - David J Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Michael J Fisher
- Children’s Hospital of Philadelphia, Section of Neuro-Oncology, Philadelphia, Pennsylvania, USA
| | - AeRang Kim
- Children’s National Hospital, Center for Cancer and Blood Disorders, Washington, District of Columbia, USA
| | - Miriam Bornhorst
- Children’s National Hospital, Center for Cancer and Blood Disorders, Washington, District of Columbia, USA
| | - Brian D Weiss
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jaishri O Blakeley
- Johns Hopkins University, Division of Neurology, Baltimore, Maryland, USA
| | - Malcolm A Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Murphy PS, Galette P, van der Aart J, Janiczek RL, Patel N, Brown AP. The role of clinical imaging in oncology drug development: progress and new challenges. Br J Radiol 2023; 96:20211126. [PMID: 37393537 PMCID: PMC10546429 DOI: 10.1259/bjr.20211126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/14/2023] [Accepted: 06/06/2023] [Indexed: 07/03/2023] Open
Abstract
In 2008, the role of clinical imaging in oncology drug development was reviewed. The review outlined where imaging was being applied and considered the diverse demands across the phases of drug development. A limited set of imaging techniques was being used, largely based on structural measures of disease evaluated using established response criteria such as response evaluation criteria in solid tumours. Beyond structure, functional tissue imaging such as dynamic contrast-enhanced MRI and metabolic measures using [18F]flourodeoxyglucose positron emission tomography were being increasingly incorporated. Specific challenges related to the implementation of imaging were outlined including standardisation of scanning across study centres and consistency of analysis and reporting. More than a decade on the needs of modern drug development are reviewed, how imaging has evolved to support new drug development demands, the potential to translate state-of-the-art methods into routine tools and what is needed to enable the effective use of this broadening clinical trial toolset. In this review, we challenge the clinical and scientific imaging community to help refine existing clinical trial methods and innovate to deliver the next generation of techniques. Strong industry-academic partnerships and pre-competitive opportunities to co-ordinate efforts will ensure imaging technologies maintain a crucial role delivering innovative medicines to treat cancer.
Collapse
Affiliation(s)
| | - Paul Galette
- Telix Pharmaceuticals (US) Inc, Fishers, United States
| | | | | | | | - Andrew P. Brown
- Vale Imaging Consultancy Solutions, Harston, Cambridge, United Kingdom
| |
Collapse
|
16
|
Pillay-Smiley N, Fletcher JS, de Blank P, Ratner N. Shedding New Light: Novel Therapies for Common Disorders in Children with Neurofibromatosis Type I. Pediatr Clin North Am 2023; 70:937-950. [PMID: 37704352 DOI: 10.1016/j.pcl.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Neurofibromatosis type I (NF1) is a common dominantly inherited disorder, and one of the most common of the RASopathies. Most individuals with NF1 develop plexiform neurofibromas and cutaneous neurofibromas, nerve tumors caused by NF1 loss of function in Schwann cells. Cell culture models and mouse models of NF1 are being used to test drug efficacy in preclinical trials, which led to Food and Drug Administration approval for use of MEK inhibitors to shrink most inoperable plexiform neurofibromas. This article details methods used for testing in preclinical models, and outlines newer models that may identify additional, curative, strategies.
Collapse
Affiliation(s)
- Natasha Pillay-Smiley
- University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jonathan S Fletcher
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Current Address: Division of Hematology-Oncology, University of Texas Southwestern, Dallas, TX, USA
| | - Peter de Blank
- University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
17
|
Hu X, Li W, Zeng K, Xu Z, Li C, Kang Z, Li S, Huang X, Han P, Lin H, Hui AM, Tan Y, Diao L, Li B, Wang X, Wu Z, Lin X. Phase 1 dose-escalation study to evaluate the safety, tolerability, pharmacokinetics, and anti-tumor activity of FCN-159 in adults with neurofibromatosis type 1-related unresectable plexiform neurofibromas. BMC Med 2023; 21:230. [PMID: 37400844 DOI: 10.1186/s12916-023-02927-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/07/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Surgery is a common treatment strategy for patients with neurofibromatosis type 1 (NF1)-related plexiform neurofibroma (PN) and has limited efficacy. FCN-159 is a novel anti-tumorigenic drug via selective inhibition of MEK1/2. This study assesses the safety and efficacy of FCN-159 in patients with NF1-related PN. METHODS This is a multicenter, open-label, single-arm, phase I dose-escalation study. Patients with NF1-related PN that was non-resectable or unsuitable for surgery were enrolled; they received FCN-159 monotherapy daily in 28-day cycles. RESULTS Nineteen adults were enrolled in the study, 3 in 4 mg, 4 in 6 mg, 8 in 8 mg, and 4 in 12 mg. Among patients included in dose-limiting toxicity (DLT) analysis, DLTs (grade 3 folliculitis) were reported in 1 of 8 patients (16.7%) receiving 8 mg and 3 of 3 (100%) patients receiving 12 mg. The maximum tolerated dose was determined to be 8 mg. FCN-159-related treatment-emergent adverse events (TEAEs) were observed in 19 patients (100%); most of which were grade 1 or 2. Nine (47.4%) patients reported grade 3 study-drug-related TEAEs across all dose levels, including four experiencing paronychia and five experiencing folliculitis. Of the 16 patients analyzed, all (100%) had reduced tumor size and six (37.5%) achieved partial responses; the largest reduction in tumor size was 84.2%. The pharmacokinetic profile was approximately linear between 4 and 12 mg, and the half-life supported once daily dosing. CONCLUSIONS FCN-159 was well tolerated up to 8 mg daily with manageable adverse events and showed promising anti-tumorigenic activity in patients with NF1-related PN, warranting further investigation in this indication. TRIAL REGISTRATION ClinicalTrials.gov, NCT04954001. Registered 08 July 2021.
Collapse
Affiliation(s)
- Xiaojie Hu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Kang Zeng
- Department of Dermatology, NanFang Hospital Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Changxing Li
- Department of Dermatology, NanFang Hospital Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Zhuang Kang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Shenglan Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Xin Huang
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Pu Han
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Hongmei Lin
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Ai-Min Hui
- Fosun Pharma USA Inc., 91 Hartwell Ave Suite 305, Lexington, MA, 02421, USA
- EnCureGen Pharma, 9 Yayingshi Road, Guangzhou, 510525, China
| | - Yan Tan
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Lei Diao
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Ben Li
- Beijing Fosun Pharmaceutical Research and Development Co., Ltd, 1289 Yishan Road, Shanghai, 200233, China
| | - Xingli Wang
- Shanghai Fosun Pharmaceutical Development Co., Ltd, 1289 Yishan Road, Shanghai, 20033, China
| | - Zhuli Wu
- Shanghai Fosun Pharmaceutical Development Co., Ltd, 1289 Yishan Road, Shanghai, 20033, China.
| | - Xiaoxi Lin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
18
|
John L, Smith H, Ilanchezhian M, Lockridge R, Reilly KM, Raygada M, Dombi E, Sandler A, Thomas BJ, Glod J, Miettinen M, Allen T, Sommer J, Levy J, Lozinsky S, Dix D, Bouffet E, MacDonald S, Mukherjee D, Snyderman CH, Rowan NR, Malyapa R, Park DM, Heery C, Gardner PA, Cote GM, Fuller S, Butman JA, Jackson S, Gulley JL, Widemann BC, Wedekind MF. The NIH pediatric/young adult chordoma clinic and natural history study: Making advances in a very rare tumor. Pediatr Blood Cancer 2023; 70:e30358. [PMID: 37347686 PMCID: PMC10739575 DOI: 10.1002/pbc.30358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/26/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Chordomas are rare tumors arising from the skull base and spine, with approximately 20 pediatric chordoma cases in the Unitedn States per year. The natural history and optimal treatment of pediatric chordomas, especially poorly differentiated and dedifferentiated subtypes, is incompletely understood. Herein, we present findings from our first National Cancer Institute (NCI) chordoma clinic and a retrospective analysis of published cases of pediatric poorly differentiated chordomas (PDC) and dedifferentiated chordomas (DC). METHODS Patients less than 40 years old with chordoma were enrolled on the NCI Natural History and Biospecimens Acquisitions Study for Children and Adults with Rare Solid Tumors protocol (NCT03739827). Chordoma experts reviewed patient records, evaluated patients, and provided treatment recommendations. Patient-reported outcomes, biospecimens, and volumetric tumor analyses were collected. A literature review for pediatric PDC and DC was conducted. RESULTS Twelve patients (median age: 14 years) attended the clinic, including four patients with active disease and three patients with PDC responsive to systemic therapy. Consensus treatment, management, and recommendations were provided to patients. Literature review returned 45 pediatric cases of PDC or DC with variable treatments and outcomes. CONCLUSIONS A multidisciplinary expert clinic was feasible and successful in improving understanding of pediatric chordoma. While multimodal approaches have all been employed, treatment for PDC has been inconsistent and a recommended standardized treatment approach has not been defined. Centralized efforts, inclusive of specialized chordoma-focused clinics, natural history studies, and prospective analyses will help in the standardization of care for this challenging disease.
Collapse
Affiliation(s)
- Liny John
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Hannah Smith
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Maran Ilanchezhian
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Robin Lockridge
- Clinical Research Directorate (CRD), Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Karlyne M Reilly
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Margarita Raygada
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Abby Sandler
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Barbara J Thomas
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - John Glod
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Markku Miettinen
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Taryn Allen
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Joan Levy
- Chordoma Foundation, Durham, NC, USA
| | | | - David Dix
- BC Children’s Hospital, Vancouver, Canada
| | | | | | | | | | | | - Robert Malyapa
- University of Maryland Medical Center, Baltimore, MD, USA
| | | | - Christopher Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Paul A. Gardner
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Sarah Fuller
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - John A. Butman
- Radiology and Imaging Sciences, The National Institutes of Health, Bethesda, MD, USA
| | - Sadhana Jackson
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - James L. Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mary Frances Wedekind
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
19
|
Kotch C, Wagner K, Broad JH, Dombi E, Minturn JE, Phillips P, Smith K, Li Y, Jacobs IN, Elden LM, Fisher MJ, Belasco J. Vinblastine/Methotrexate for Debilitating and Progressive Plexiform Neurofibroma in Children and Young Adults with Neurofibromatosis Type 1: A Phase 2 Study. Cancers (Basel) 2023; 15:cancers15092621. [PMID: 37174087 PMCID: PMC10177272 DOI: 10.3390/cancers15092621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Limited therapies exist for neurofibromatosis type 1 (NF1)-associated plexiform neurofibroma (PN). For this reason, the activity of vinblastine (VBL) and methotrexate (MTX) was evaluated in children and young adults with NF1 and PN. Patients ≤ 25 years of age with progressive and/or inoperable NF1-PN received VBL 6 mg/m2 and MTX 30 mg/m2 weekly for 26 weeks, followed by every 2 weeks for 26 weeks. Objective response rate was the primary endpoint. Of 25 participants enrolled, 23 were evaluable. The median age of participants was 6.6 years (range 0.3-20.7). The most frequent toxicities were neutropenia and elevation of transaminases. On two-dimensional (2D) imaging, 20 participants (87%) had stable tumor, with a median time to progression of 41.5 months (95% confidence interval 16.9, 64.9). Two of eight participants (25%) with airway involvement demonstrated functional improvements including decreased positive pressure requirements and apnea-hypopnea index. A post hoc three-dimensional (3D) analysis of PN volumes was completed on 15 participants with amenable imaging; 7 participants (46%) had progressive disease on or by the end of therapy. VBL/MTX was well-tolerated but did not result in objective volumetric response. Furthermore, 3D volumetric analysis highlighted the lack of sensitivity of 2D imaging for PN response evaluation.
Collapse
Affiliation(s)
- Chelsea Kotch
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristina Wagner
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - J Harris Broad
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Anesthesiology, Valley Medical Center, Renton, WA 98055, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jane E Minturn
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Phillips
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine Smith
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yimei Li
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian N Jacobs
- Division of Otolaryngology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lisa M Elden
- Division of Otolaryngology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Michael J Fisher
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean Belasco
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Welling DB. Targeted Therapies in the Treatment of Vestibular Schwannomas: Current State and New Horizons. Otolaryngol Clin North Am 2023; 56:543-556. [PMID: 37024334 DOI: 10.1016/j.otc.2023.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Vestibular schwannomas continue to cause hearing loss, facial nerve paralysis, imbalance, and tinnitus. These symptoms are compounded by germline neurofibromatosis type 2 (NF2) gene loss and multiple intracranial and spinal cord tumors associated with NF2-related schwannomatosis. The current treatments of observation, microsurgical resection, or stereotactic radiation may prevent catastrophic brainstem compression but are all associated with the loss of cranial nerve function, particularly hearing loss. Novel targeted treatment options to stop tumor progression include small molecule inhibitors, immunotherapy, anti-inflammatory drugs, radio-sensitizing and sclerosing agents, and gene therapy.
Collapse
Affiliation(s)
- D Bradley Welling
- Harvard Department of Otolaryngology Head & Neck Surgery, 243 Charles Street, Boston, MA, USA; Massachusetts Eye and Ear Infirmary and Massachusetts General Hospital.
| |
Collapse
|
21
|
Wataya-Kaneda M, Watanabe Y, Nakamura A, Yamamoto K, Okada K, Maeda S, Nimura K, Saga K, Katayama I. Pilot study for the treatment of cutaneous neurofibromas in neurofibromatosis type 1 patients using topical sirolimus gel. J Am Acad Dermatol 2023; 88:877-880. [PMID: 36334988 DOI: 10.1016/j.jaad.2022.08.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Mari Wataya-Kaneda
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Neurocutaneous Medicine, Division of Health Sciences, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Yoshiyuki Watanabe
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Radiology, Shiga University of Medical Science, Otsu, Japan
| | - Ayumi Nakamura
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Kouji Yamamoto
- Department of Biostatistics Yokohama City University, School of Medicine, Yokohama, Japan
| | - Kiyoshi Okada
- Strategic Global Partnership Cross-Innovation Initiative, Osaka University Graduate School of Medicine, Faculty of Medicine, Osaka University Hospital, Suita, Japan
| | - Shinichiro Maeda
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kotaro Saga
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ichiro Katayama
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
22
|
Douwes JPJ, Koetsier KS, van Dam VS, Plotkin SR, Barker FG, Welling DB, Jansen JC, Hensen EF, Shih HA. Proton Radiotherapy for Vestibular Schwannomas in Patients with NF2-Related Schwannomatosis: A Case Series. Curr Oncol 2023; 30:3473-3483. [PMID: 36975476 PMCID: PMC10047060 DOI: 10.3390/curroncol30030263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
(1) Background: This study aimed to evaluate the efficacy and treatment-related toxicity of proton radiotherapy (PRT) for vestibular schwannoma (VS) in patients with neurofibromatosis type 2-related schwannomatosis (NF2). (2) Methods: Consecutive NF2 patients treated with PRT for VS between 2004 and 2016 were retrospectively evaluated, focusing on tumor volume, facial and trigeminal nerve function, hearing, tinnitus, vestibular symptoms, and the need for salvage therapy after PRT. (3) Results: Eight patients were included (median age 36 years, 50% female). Median follow-up was 71 months. Five (63%) patients received fractionated PRT and three (38%) received PRT radiosurgery for VS. Six patients (75%) received prior VS surgery; three also received bevacizumab. Six patients (75%) did not require salvage therapy after PRT. Two patients (25%) with residual hearing lost it after PRT, and six had already lost ipsilateral hearing prior to PRT. Tumor and treatment-related morbidity could be evaluated in six patients. Following PRT, conditions that occurred or worsened were: facial paresis in five (83%), trigeminal hypoesthesia in two (33%), tinnitus in two (33%), and vestibular symptoms in four patients (67%). (4) Conclusion: After PRT for VS, the majority of the NF2 patients in the cohort did not require additional therapy. Tumor and/or treatment-related cranial nerve deficits were common. This is at least partly explained by the use of PRT as a salvage treatment after surgery or bevacizumab, in the majority of cases. There remains the further opportunity to elucidate the efficacy and toxicity of proton radiotherapy as a primary treatment.
Collapse
Affiliation(s)
- Jules P J Douwes
- Department of Otorhinolaryngology-Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Kimberley S Koetsier
- Department of Otorhinolaryngology-Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Victor S van Dam
- Department of Otorhinolaryngology-Head and Neck Surgery, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Scott R Plotkin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Frederick G Barker
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - D Bradley Welling
- Department of Otorhinolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Jeroen C Jansen
- Department of Otorhinolaryngology-Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Erik F Hensen
- Department of Otorhinolaryngology-Head and Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Helen A Shih
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
23
|
Ly KI, Merker VL, Cai W, Bredella MA, Muzikansky A, Thalheimer RD, Da JL, Orr CC, Herr HP, Morris ME, Chang CY, Harris GJ, Plotkin SR, Jordan JT. Ten-Year Follow-up of Internal Neurofibroma Growth Behavior in Adult Patients With Neurofibromatosis Type 1 Using Whole-Body MRI. Neurology 2023; 100:e661-e670. [PMID: 36332985 PMCID: PMC9969927 DOI: 10.1212/wnl.0000000000201535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Internal neurofibromas, including plexiform neurofibromas (PNF), can cause significant morbidity in patients with neurofibromatosis type 1 (NF1). PNF growth is most pronounced in children and young adults, with more rapid growth thought to occur in a subset of PNF termed distinct nodular lesions (DNL). Growth behavior of internal neurofibromas and DNL in older adults is not well documented; yet knowledge thereof is important for patient risk stratification and clinical trial design. The primary objective of this study was to evaluate the long-term growth behavior of internal neurofibromas in adults with NF1. Secondary objectives were to correlate tumor growth behavior with patient-specific, tumor-specific, and patient-reported variables. METHODS In this prospective cohort study, internal neurofibromas were identified on coronal short TI inversion recovery sequences on baseline and follow-up whole-body MRIs (WBMRIs). Tumor growth and shrinkage were defined as a volume change ≥20%. The association between tumor growth and patient-specific (baseline age, sex, and genotype), tumor-specific (morphology, location, DNL presence on baseline WBMRI, and maximum standardized uptake value on baseline PET imaging), and patient-reported variables (endogenous and exogenous hormone exposure, pain intensity, and quality of life) was assessed using the Spearman correlation coefficient and Kruskal-Wallis test. RESULTS Of 106 patients with a baseline WBMRI obtained as part of a previous research study, 44 had a follow-up WBMRI. Three additional patients with WBMRIs acquired for clinical care were included, generating 47 adults for this study. The median age during baseline WBMRI was 42 years (range 18-70). The median time between WBMRIs was 10.4 years. Among 324 internal neurofibromas, 62.8% (56% of PNF and 62.1% of DNL) shrank spontaneously without treatment and 17.1% (17.9% of PNF and 13.8% of DNL) grew. Growth patterns were heterogeneous within participants. Patient-specific, tumor-specific, and patient-reported variables (including endogenous and exogenous hormone exposure) were not strong predictors of tumor growth. DISCUSSION Internal neurofibroma growth behavior in older adults differs fundamentally from that in children and young adults, with most tumors, including DNL, demonstrating spontaneous shrinkage. Better growth models are needed to understand factors that influence tumor growth. These results will inform clinical trial design for internal neurofibromas.
Collapse
Affiliation(s)
- K Ina Ly
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston.
| | - Vanessa L Merker
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Wenli Cai
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Miriam A Bredella
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Alona Muzikansky
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Raquel D Thalheimer
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Jennifer Liwei Da
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Christina C Orr
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Hamilton P Herr
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Mary E Morris
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Connie Y Chang
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Gordon J Harris
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Scott R Plotkin
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| | - Justin T Jordan
- From the Stephen E. and Catherine Pappas Center for Neuro-Oncology (K.I.L., V.L.M., R.D.T., J.L.D., C.C.O., H.P.H., S.R.P., J.T.J.), Massachusetts General Hospital; Department of Radiology (W.C., M.A.B., C.Y.C., G.J.H.), Massachusetts General Hospital; Biostatistics Center (A.M.), Massachusetts General Hospital; and Department of Obstetrics and Gynecology (M.E.M.), Massachusetts General Hospital, Boston
| |
Collapse
|
24
|
Efficacy Endpoints in Phase II Clinical Trials for Meningioma: An Analysis of Recent Clinical Trials. Ther Innov Regul Sci 2023; 57:603-610. [PMID: 36602756 DOI: 10.1007/s43441-022-00494-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Response Evaluation Criteria in Solid Tumors (RECIST)-based response rates are commonly used as efficacy endpoints in phase II clinical trials for solid tumors. However, no consensus has been reached concerning adequate efficacy endpoints for phase II clinical trials targeting meningioma. Irregularity of lesions after resection, and varying degrees of dysplasia and histologic subtypes make establishing an appropriate efficacy evaluation difficult. METHODS We analyzed primary efficacy endpoints (PEEs) and background factors from 48 trials retrieved from ClinicalTrials.gov ( https://clinicaltrials.gov/ ) using the search criteria "meningioma," "interventional," "phase II," and "study start 4/1/2001 to 3/31/2021." Primary purpose of the study was efficacy endpoint setting in overall population and three subgroups. RESULTS Among 45 PEEs set in the 39 trials included; 33 trials with single PEE, and six trials with double PEEs, 17/45 (38%) trials adopted progression-free survival (PFS) rate, 15/45 (33%) trials response rate (seven Macdonald criteria or modified, three RECIST, three volumetric estimation, one RANO criteria, one unknown), 10/45 (22%) PFS, 1/45 (2%) OS, and 2/45 (4%) other endpoints. Although 26 PEEs were time-to-event endpoints, 19 of the 26 PEEs were single-arm studies. CONCLUSIONS Time-to-event efficacy endpoints were often compared to historical data, and two-dimensional evaluation is more suitable than one-dimensional one. Accumulation of prognostic data is essential to standardize time-to-event efficacy endpoints. Considering the difficulty of setting design for phase II clinical studies targeting meningioma, evaluation might be done with multiple efficacy endpoints.
Collapse
|
25
|
Cacchione A, Fabozzi F, Carai A, Colafati GS, del Baldo G, Rossi S, Diana M, Megaro G, Milano GM, Macchiaiolo M, Crocoli A, De Ioris MA, Boccuto L, Secco DE, Zama M, Agolini E, Tomà P, Mastronuzzi A. Safety and Efficacy of Mek Inhibitors in the Treatment of Plexiform Neurofibromas: A Retrospective Study. Cancer Control 2023; 30:10732748221144930. [PMID: 36598023 PMCID: PMC9830579 DOI: 10.1177/10732748221144930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Plexiform neurofibromas (PN) represent the main cause of morbidity in patients affected by Neurofibromatosis Type 1 (NF1). Until recently, surgery has been the main treatment option in these patients, but it is burdened with a low efficacy rate and a high incidence of side effects as well as recurrence. In recent years, MEK inhibitors (MEKi) such as selumetinib and trametinib have shown great promise. METHODS We retrospectively describe a single center cohort of NF1 patients affected by PN1 and treated with MEKi since 2019 to 2021. Patients recruited in the study were affected by PN that were not eligible to complete surgical excision, symptomatic or with major cosmetic deformation or functional neurological deficits. RESULTS Most patients experienced improvement in clinical symptoms and quality of life, with reduction or stabilization of lesions. However, no complete response was achieved. The most common adverse effects involved the skin, affecting every patient. Importantly, no life-threatening adverse effects occurred. CONCLUSIONS In our experience, MEKi treatment has been shown to be both safe and effective in improving symptomatology and quality of life.
Collapse
Affiliation(s)
- Antonella Cacchione
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Francesco Fabozzi
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy,Department of Pediatrics, University of Tor Vergata, Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | - Giada del Baldo
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Sabrina Rossi
- Pathology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Martino Diana
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Giacomina Megaro
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Giuseppe Maria Milano
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, Academic Department of Pediatrics, Bambino Gesù Children’s Hospital, Rome, Italy
| | | | - Maria Antonietta De Ioris
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Luigi Boccuto
- Healthcare Genetics Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, USA
| | - Domitilla Elena Secco
- PsD of Department of Paediatric Haematology/Oncology, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Mario Zama
- Surgery Department, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Emanuele Agolini
- Laboratory of Medical Genetics, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Paolo Tomà
- Department of Imaging, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - Angela Mastronuzzi
- Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome, Italy,Unicamillus, Saint Camillus International University of Health Sciences, Rome, Italy,Angela Mastronuzzi, MD, PhD, Department of Hematology/Oncology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children’s Hospital, Rome 00165, Italy.
| |
Collapse
|
26
|
Jordan JT, Orr CC, Thalheimer RD, Cambillo JV, Beauchamp RL, Shaikh G, Muzikansky A, Stemmer-Rachamimov A, Giovannini M, Kalamarides M, Barker FG, Ramesh V, Plotkin SR. Prospective phase II trial of the dual mTORC1/2 inhibitor vistusertib for progressive or symptomatic meningiomas in persons with neurofibromatosis 2. Neurooncol Adv 2023; 5:vdad041. [PMID: 37215956 PMCID: PMC10195194 DOI: 10.1093/noajnl/vdad041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Background Meningiomas occur in 80% of persons with neurofibromatosis 2 (NF2) and cause significant mortality and morbidity, yet there are no effective medical treatments. NF2-deficient tumors have constitutive activation of mammalian/mechanistic target of rapamycin (mTOR), and treatment with mTORC1 inhibitors results in growth arrest in a minority of tumors, with paradoxical activation of the mTORC2/AKT pathway. We studied the effect of vistusertib, a dual mTORC1/mTORC2 inhibitor, in NF2 patients with progressive or symptomatic meningiomas. Methods Vistusertib was administered orally at 125 mg twice daily for 2 consecutive days each week. The primary endpoint was the imaging response in the target meningioma, defined as a volume decrease of 20% compared with the baseline. Secondary endpoints included toxicity, imaging response of nontarget tumors, quality of life, and genetic biomarkers. Results Eighteen participants (13 female), median age of 41 (range, 18-61) years, were enrolled. In target meningiomas, the best response was partial response (PR) in 1/18 tumors (6%) and stable disease (SD) in 17/18 tumors (94%). For all measured intracranial meningiomas and vestibular schwannomas, the best imaging response was PR in 6/59 tumors (10%) and SD in 53 (90%). Treatment-related grade 3/4 adverse events occurred in 14 (78%) participants, and 9 participants discontinued treatment due to side effects. Conclusions Although the study did not meet the primary endpoint, vistusertib treatment was associated with high rates of SD in progressive NF2-related tumors. However, this dosing regimen for vistusertib was poorly tolerated. Future studies of dual mTORC inhibitors for NF2 should focus on optimizing tolerability and evaluating the relevance of tumor stability in participants.
Collapse
Affiliation(s)
- Justin T Jordan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christina C Orr
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raquel D Thalheimer
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Josephine V Cambillo
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Roberta L Beauchamp
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ghalib Shaikh
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alona Muzikansky
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anat Stemmer-Rachamimov
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, Los Angeles, CA, USA
| | - Michel Kalamarides
- Department of Neurosurgery, Hopital Pitie-Salpetriere, Sorbonne Université, Paris, France
| | - Fred G Barker
- Neurosurgical Service, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vijaya Ramesh
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Scott R Plotkin
- Corresponding Author: Scott R. Plotkin, MD, PhD, Department of Neurology, Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Yawkey 9E, Boston, MA 02114, USA ()
| |
Collapse
|
27
|
Paterra R, Bettinaglio P, Borghi A, Mangano E, Tritto V, Cesaretti C, Schettino C, Bordoni R, Santoro C, Avignone S, Moscatelli M, Melone MAB, Saletti V, Piluso G, Natacci F, Riva P, Eoli M. A Translational Approach to Spinal Neurofibromatosis: Clinical and Molecular Insights from a Wide Italian Cohort. Cancers (Basel) 2022; 15:cancers15010059. [PMID: 36612057 PMCID: PMC9817775 DOI: 10.3390/cancers15010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Spinal neurofibromatosis (SNF), a phenotypic subclass of neurofibromatosis 1 (NF1), is characterized by bilateral neurofibromas involving all spinal roots. In order to deepen the understanding of SNF’s clinical and genetic features, we identified 81 patients with SNF, 55 from unrelated families, and 26 belonging to 19 families with at least 1 member affected by SNF, and 106 NF1 patients aged >30 years without spinal tumors. A comprehensive NF1 mutation screening was performed using NGS panels, including NF1 and several RAS pathway genes. The main features of the SNF subjects were a higher number of internal neurofibromas (p < 0.001), nerve root swelling (p < 0.001), and subcutaneous neurofibromas (p = 0.03), while hyperpigmentation signs were significantly less frequent compared with the classical NF1-affected cohorts (p = 0.012). Fifteen patients underwent neurosurgical intervention. The histological findings revealed neurofibromas in 13 patients and ganglioneuromas in 2 patients. Phenotypic variability within SNF families was observed. The proportion of missense mutations was higher in the SNF cases than in the classical NF1 group (21.40% vs. 7.5%, p = 0.007), conferring an odds ratio (OR) of 3.34 (CI = 1.33−10.78). Two unrelated familial SNF cases harbored in trans double NF1 mutations that seemed to have a subclinical worsening effect on the clinical phenotype. Our study, with the largest series of SNF patients reported to date, better defines the clinical and genetic features of SNF, which could improve the management and genetic counseling of NF1.
Collapse
Affiliation(s)
- Rosina Paterra
- Molecular Neuroncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20122 Milano, Italy
| | - Paola Bettinaglio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Segrate, 20122 Milan, Italy
| | - Arianna Borghi
- Molecular Neuroncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20122 Milano, Italy
| | - Eleonora Mangano
- Institute of Biomedical Technologies (ITB) National Research Center (CNR), ITB-CNR, Segrate, 20122 Milan, Italy
| | - Viviana Tritto
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Segrate, 20122 Milan, Italy
| | - Claudia Cesaretti
- Medical Genetics Unit, Woman-Child-Newborn Department, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Carla Schettino
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy
| | - Roberta Bordoni
- Institute of Biomedical Technologies (ITB) National Research Center (CNR), ITB-CNR, Segrate, 20122 Milan, Italy
| | - Claudia Santoro
- Neurofibromatosis Referral Center, Department of Woman, Child and of General and Specialized Surgery, and Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy
| | - Sabrina Avignone
- Neuroradiology Department, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Marco Moscatelli
- Neuroradiology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milano, Italy
| | - Mariarosa Anna Beatrice Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA
| | - Veronica Saletti
- Developmental Neurology Unit, Mariani Foundation Center for Complex Disabilities, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Giulio Piluso
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Naples, Italy
| | - Federica Natacci
- Medical Genetics Unit, Woman-Child-Newborn Department, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Paola Riva
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Segrate, 20122 Milan, Italy
- Correspondence: (P.R.); (M.E.); Tel.: +39-02-503130462 (P.R.); +39-02-23942285 (M.E.)
| | - Marica Eoli
- Molecular Neuroncology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20122 Milano, Italy
- Correspondence: (P.R.); (M.E.); Tel.: +39-02-503130462 (P.R.); +39-02-23942285 (M.E.)
| |
Collapse
|
28
|
Fleming J, Morgan O, Wong C, Schlub TE, Berman Y. Characterization of health concerns in people with neurofibromatosis type 1. Mol Genet Genomic Med 2022; 11:e2077. [PMID: 36444392 PMCID: PMC9834143 DOI: 10.1002/mgg3.2077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Neurofibromatosis 1 (NF1) is a common cancer predisposition syndrome. Affected individuals require lifelong surveillance and often suffer progressive disfigurement due to cutaneous neurofibromas. The aim of this research was to characterize health concerns and quality of life (QOL) in a population cohort. METHODS An online survey was completed by 68 adults and 32 parents of children with NF1, and 60 controls. The survey included the Skindex-29 QOL scale, 5D-itch scale, and additional health questions. RESULTS Frequency of itch was high in children (50%) and adults (69%), with most expressing interest in treatment for itch. The presence of itch and increased visibility of NF1 were predictors of poorer QoL. Many adults (53%) and parents (44%) desired access to treatment to improve cosmetic appearance. Muscle weakness/tiredness was also prevalent amongst (60-70%) adults and children with NF1. Two-thirds of adults with NF1 reported limited awareness of NF1 services and poor knowledge of surveillance, particularly breast screening in young women. CONCLUSION This study highlights the impact of NF1-related itch and visibility in adults and children with a need for cosmetic and itch treatment. The findings emphasize a need for strategies to promote awareness, and access to management and treatment of NF1 in adults.
Collapse
Affiliation(s)
- Jane Fleming
- Department of Clinical GeneticsNorthern Sydney Local Health DistrictSydneyNew South WalesAustralia
| | - Oliver Morgan
- Faculty of Health and MedicineUniversity of Sydney, Northern Clinical SchoolSydneyNew South WalesAustralia
| | - Claire Wong
- Department of Clinical GeneticsNorthern Sydney Local Health DistrictSydneyNew South WalesAustralia,Department of Clinical GeneticsThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Timothy E. Schlub
- Sydney School of Public Health, Faculty of Medicine and HealthUniversity of SydneyCamperdownNew South WalesAustralia
| | - Yemima Berman
- Department of Clinical GeneticsNorthern Sydney Local Health DistrictSydneyNew South WalesAustralia
| |
Collapse
|
29
|
Fisher MJ, Blakeley JO, Weiss BD, Dombi E, Ahlawat S, Akshintala S, Belzberg AJ, Bornhorst M, Bredella MA, Cai W, Ferner RE, Gross AM, Harris GJ, Listernick R, Ly I, Martin S, Mautner VF, Salamon JM, Salerno KE, Spinner RJ, Staedtke V, Ullrich NJ, Upadhyaya M, Wolters PL, Yohay K, Widemann BC. Management of neurofibromatosis type 1-associated plexiform neurofibromas. Neuro Oncol 2022; 24:1827-1844. [PMID: 35657359 PMCID: PMC9629437 DOI: 10.1093/neuonc/noac146] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Plexiform Neurofibromas (PN) are a common manifestation of the genetic disorder neurofibromatosis type 1 (NF1). These benign nerve sheath tumors often cause significant morbidity, with treatment options limited historically to surgery. There have been tremendous advances over the past two decades in our understanding of PN, and the recent regulatory approvals of the MEK inhibitor selumetinib are reshaping the landscape for PN management. At present, there is no agreed upon PN definition, diagnostic evaluation, surveillance strategy, or clear indications for when to initiate treatment and selection of treatment modality. In this review, we address these questions via consensus recommendations from a panel of multidisciplinary NF1 experts.
Collapse
Affiliation(s)
- Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jaishri O Blakeley
- Division of Neuro-Oncology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brian D Weiss
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Eva Dombi
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Shivani Ahlawat
- Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Allan J Belzberg
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Miriam Bornhorst
- Family Neurofibromatosis Institute, Center for Neuroscience and Behavioral Medicine,Children's National Hospital, Washington, District of Columbia, USA
| | - Miriam A Bredella
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wenli Cai
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rosalie E Ferner
- Neurofibromatosis Service, Department of Neurology, Guy's Hospital, Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Andrea M Gross
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Gordon J Harris
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Listernick
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Staci Martin
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Victor F Mautner
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes M Salamon
- Department for Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kilian E Salerno
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Verena Staedtke
- Division of Neuro-Oncology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Meena Upadhyaya
- Division of Cancer and Genetics, Cardiff University, Wales, UK
| | - Pamela L Wolters
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Kaleb Yohay
- Grossman School of Medicine, Department of Neurology, New York, New York, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Dabhi N, Pikis S, Mantziaris G, Tripathi M, Warnick R, Peker S, Samanci Y, Berger A, Bernstein K, Kondziolka D, Niranjan A, Lunsford LD, Sheehan JP. Stereotactic radiosurgery for the treatment of hypoglossal schwannoma: a multi-institutional retrospective study. Acta Neurochir (Wien) 2022; 164:2473-2481. [PMID: 35347448 DOI: 10.1007/s00701-022-05187-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Surgical removal has been performed as the first line treatment for symptomatic or enlarging hypoglossal schwannomas (HS). Stereotactic radiosurgery (SRS) offers a minimally invasive approach that may afford long-term tumor control for patients with HS particularly those who refuse or are unfit for surgery. This study evaluates outcomes after SRS performed for both newly diagnosed and residual tumors after incomplete resection. METHODS This retrospective, multi-institutional study involved patients treated with adjuvant or primary SRS for HS. The study end-points included local tumor response, clinical outcomes, and procedure-related complications. All the patients had Gamma Knife SRS. RESULTS The cohort included 12 patients (five females), median age at SRS 49.5 years (range, 37-76)]. The median tumor target volume was 5.9 cm3 (range, 0.7-27.23). At median imaging follow-up of 37 months (range, 6-153), tumor control was achieved in 11 patients. Tumor enlargement that was managed with surgical resection was noted at the 6-month follow-up in one patient. At median clinical follow-up of 30.5 months (range, 6-157), stability, or improvement of all pre-SRS signs and symptoms was noted in nine patients. Two patients experienced worsening of at least one pre-existing symptoms or sign. New-onset trapezius weakness was noted in one patient and tongue atrophy in two patients. CONCLUSION Single-fraction SRS appears to be a safe and effective upfront and adjuvant treatment option for HS. SRS may be recommended as an alternative to surgery for patients presenting with HS or as an adjuvant treatment following subtotal resection and at HS recurrence.
Collapse
Affiliation(s)
- Nisha Dabhi
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Stylianos Pikis
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Georgios Mantziaris
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Manjul Tripathi
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ronald Warnick
- Gamma Knife Center, The Jewish Hospital, Cincinnati, OH, USA
| | - Selcuk Peker
- Department of Neurosurgery, Koc University School of Medicine, Istanbul, Turkey
| | - Yavuz Samanci
- Department of Neurosurgery, Koc University School of Medicine, Istanbul, Turkey
| | - Assaf Berger
- Department of Neurosurgery, New York University Langone, New York, NY, USA
| | - Kenneth Bernstein
- Department of Radiation Oncology, New York University Langone, New York, NY, USA
| | - Douglas Kondziolka
- Department of Neurosurgery, New York University Langone, New York, NY, USA
| | - Ajay Niranjan
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - L Dade Lunsford
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
31
|
Wang D, Ge L, Guo Z, Li Y, Zhu B, Wang W, Wei C, Li Q, Wang Z. Efficacy and Safety of Trametinib in Neurofibromatosis Type 1-Associated Plexiform Neurofibroma and Low-Grade Glioma: A Systematic Review and Meta-Analysis. Pharmaceuticals (Basel) 2022; 15:956. [PMID: 36015104 PMCID: PMC9415905 DOI: 10.3390/ph15080956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 02/01/2023] Open
Abstract
Trametinib has been used in neurofibromatosis type 1 (NF1) patients, especially those with unresectable nerve tumors, but no systematic review based on the latest studies has been published. We conducted this meta-analysis to evaluate the effectiveness and safety of trametinib in treating NF1-related nerve tumors. Original articles reporting the efficacy and safety of trametinib in NF1 patents were identified in PubMed, EMBASE, and Web of Science up to 1 June 2022. Using R software and the 'meta' package, the objective response rates (ORRs) and disease control rates (DCRs) were calculated to evaluate the efficacy, and the pooled proportion of adverse events (AEs) was calculated. The Grading of Recommendations, Assessment, Development and Evaluation system was used to assess the quality of evidence. Eight studies involving 92 patients were included, which had a very low to moderate quality of evidence. The pooled ORR was 45.3% (95% CI: 28.9-62.1%, I2 = 0%), and the DCR was 99.8% (95% CI: 95.5-100%, I2 = 0%). The most common AEs was paronychia, with a pooled rate of 60.7% (95% CI: 48.8-72.7%, I2 = 0%). Our results indicate the satisfactory ability to stabilize tumor progression but a more limited ability to shrink tumors of trametinib in NF1-related nerve tumors. The safety profile of trametinib is satisfactory.
Collapse
Affiliation(s)
- Dun Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, No 37 Wainan Guoxue Road, Chengdu 610041, China
| | - Lingling Ge
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Zizhen Guo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Beiyao Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| | - Zhichao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (D.W.); (L.G.); gzzhen2016-@sjtu.edu.cn (Z.G.); (Y.L.); (B.Z.); (W.W.); (C.W.)
| |
Collapse
|
32
|
Vaassen P, Dürr NR, Rosenbaum T. Treatment of Plexiform Neurofibromas with MEK Inhibitors: First Results with a New Therapeutic Option. Neuropediatrics 2022; 53:52-60. [PMID: 34905788 DOI: 10.1055/s-0041-1740549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Neurofibromatosis type-1 (NF1)-associated plexiform neurofibromas (PN) are peripheral nerve sheath tumors that can significantly affect the quality of life. Until recently, surgery was the only treatment for these tumors. However, in most cases, surgery cannot achieve complete tumor removal and carries a high risk of postoperative deficits. Therefore, the recent approval of the MEK inhibitor selumetinib for the treatment of NF1-associated PN provides a long-awaited novel therapeutic option. Here, we report our experience with MEK inhibitor treatment in 12 pediatric NF1 patients with inoperable symptomatic PN. Eight patients received trametinib (median therapy duration 12.13 months and range 4-29 months), and four patients received selumetinib (median therapy duration 6.25 months and range 4-11 months). Volumetric magnetic resonance imaging (MRI) after 6 months of treatment was available for seven trametinib patients (median tumor volume reduction of 26.5% and range 11.3-55.7%) and two selumetinib patients (21.3% tumor volume reduction in one patient and +3% tumor volume change in the other one). All patients reported clinical benefits such as improved range of motion or reduced disfigurement. Therapy-related adverse events occurred in 58.3% of patients and mainly consisted of skin toxicity, paronychia, and gastrointestinal symptoms. Two patients discontinued trametinib treatment after 14 and 29 months when severe skin toxicity occurred and no further reduction of tumor size was observed. In one patient, discontinuation of therapy resulted in a 27.2% tumor volume increase as demonstrated on volumetric MRI 6 months later. Our data show that MEK inhibition is a novel therapeutic approach for inoperable PN with promising results and a manageable safety profile.
Collapse
Affiliation(s)
- Pia Vaassen
- Department of Pediatrics, Sana Kliniken Duisburg, Duisburg, Germany
| | | | | |
Collapse
|
33
|
Hwang J, Yoon HM, Lee BH, Kim PH, Kim KW. Efficacy and Safety of Selumetinib in Pediatric Patients With Neurofibromatosis Type 1: A Systematic Review and Meta-analysis. Neurology 2022; 98:e938-e946. [PMID: 35017312 DOI: 10.1212/wnl.0000000000013296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/27/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Although the recent approval of selumetinib is expected to transform the management of children with Neurofibromatosis type 1 (NF1), particularly those with symptomatic and inoperable PN, no systematic review has summarized their efficacy and safety based on the latest studies. This study was conducted to systematically evaluate the efficacy and safety of selumetinib in children with NF1 METHODS: Original articles reporting the efficacy and safety of selumetinib in patients with NF1 were identified in PubMed and EMBASE up to January 28, 2021. The pooled objective response rates (ORRs) and disease control rates (DCRs) were calculated using the DerSimonian-Laird method based on random-effects modeling. The pooled proportion of adverse events (AEs) was also calculated. The quality of the evidence was assessed using the Grading of Recommendations, Assessment, Development and Evaluation system. RESULTS Five studies involving 126 patients were included in our analysis. The studies had a very low to moderate quality of the evidence. The pooled ORR was 73.8% (95% CI: 57.3-85.5%), and the DCR was 92.5% (95% CI: 66.5-98.7%). The two most common AEs were diarrhea, which had a pooled rate of 63.8% (95% CI, 52.9-73.4%) and an increase in creatine kinase levels, which had a pooled rate of 63.3% (95% CI, 35.6-84.3%). DISCUSSION Our results indicate that selumetinib is an effective and safe treatment for pediatric patients with symptomatic, inoperable plexiform neurofibromas. Further larger-scale randomized controlled studies are needed to confirm the long-term outcome of patients treated with this drug.
Collapse
Affiliation(s)
- Jisun Hwang
- Department of Radiology, Dongtan Sacred Heart Hospital, Hallym University Medical Center, 7, Keunjaebong-gil, Hwaseong-si, Gyeonggi-do 18450, Republic of Korea
| | - Hee Mang Yoon
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Pyeong Hwa Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Kyung Won Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| |
Collapse
|
34
|
Kilian A, Aigner A, Simon M, Salchow DJ, Potratz C, Thomale UW, Hernáiz Driever P, Tietze A. Tumor load rather than contrast enhancement is associated with the visual function of children and adolescents with optic pathway glioma - a retrospective Magnetic Resonance Imaging study. J Neurooncol 2022; 156:589-597. [PMID: 34994964 PMCID: PMC8860805 DOI: 10.1007/s11060-021-03941-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/29/2021] [Indexed: 11/06/2022]
Abstract
Introduction Optic pathway gliomas are often asymptomatic tumors occurring in children with neurofibromatosis type 1 (NF1 + OPG) or sporadically (spOPG). Treatment is usually prompted by visual loss and/or tumor progression on MRI. The aim of this study was to investigate the relationship between visual acuity (VA), tumor growth, and contrast enhancement to provide more distinct indications for the administration of gadolinium-based contrast agents. Methods Tumor load was retrospectively measured and enhancement semi-quantitatively scored on 298 MRIs of 35 patients (63% NF1 + OPG). Spearman rank correlation between tumor load and enhancement was calculated and a linear mixed model used to examine the influence of tumor load and enhancement on corresponding VA tests (LogMAR). Results The optic nerve width in NF1 + OPGs was strongly associated with VA (regression coefficient 0.75; confidence interval 0.61—0.88), but weakly with enhancement (0.06; −0.04—0.15). In spOPGs, tumor volume and optic nerve width were more relevant (0.31; −0.19—0.81 and 0.39; 0.05—0.73) than enhancement (0.09; −0.09—0.27). Conclusions Tumor load measures may be more relevant for the surveillance of optic pathway gliomas than enhancement, given that VA is the relevant outcome parameter. Regular contrast administration should therefore be questioned in these patients. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03941-1.
Collapse
Affiliation(s)
- Anna Kilian
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neuroradiology, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Annette Aigner
- Charité - Universitätsmedizin Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| | - Michèle Simon
- Charité - Universitätsmedizin Berlin, Department of Pediatric Oncology and Hematology, Berlin, Germany
| | - Daniel J Salchow
- Charité - Universitätsmedizin Berlin, Department of Ophthalmology, Berlin, Germany
| | - Cornelia Potratz
- Charité - Universitätsmedizin Berlin, Department of Pediatric Neurology, Berlin, Germany
| | - Ulrich-Wilhelm Thomale
- Charité - Universitätsmedizin Berlin, Department of Pediatric Neurosurgery, Berlin, Germany
| | - Pablo Hernáiz Driever
- Charité - Universitätsmedizin Berlin, Department of Pediatric Oncology and Hematology, Berlin, Germany.,German HIT-LOGGIC-Registry for Children and Adolescents With Low-Grade Glioma, Berlin, Germany
| | - Anna Tietze
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neuroradiology, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
35
|
Gross AM, Glassberg B, Wolters PL, Dombi E, Baldwin A, Fisher MJ, Kim A, Bornhorst M, Weiss BD, Blakeley JO, Whitcomb P, Paul SM, Steinberg SM, Venzon DJ, Martin S, Carbonell A, Heisey K, Therrien J, Kapustina O, Dufek A, Derdak J, Smith MA, Widemann BC. OUP accepted manuscript. Neuro Oncol 2022; 24:1978-1988. [PMID: 35467749 PMCID: PMC9629448 DOI: 10.1093/neuonc/noac109] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Selumetinib was recently approved for the treatment of inoperable symptomatic plexiform neurofibromas (PNs) in children with neurofibromatosis type 1 (NF1). This parallel phase II study determined the response rate to selumetinib in children with NF1 PN without clinically significant morbidity. METHODS Children with NF1 and inoperable PNs, which were not yet causing clinically significant morbidity but had the potential to cause symptoms, received selumetinib at 25 mg/m2 orally twice daily (1 cycle = 28 days). Volumetric magnetic resonance imaging analysis and outcome assessments, including patient-reported (PRO), observer-reported, and functional outcome measures were performed every 4 cycles for 2 years, with changes assessed over time. A confirmed partial response (cPR) was defined as PN volume decrease of ≥20% on at least 2 consecutive scans ≥3 months apart. RESULTS 72% of subjects experienced a cPR on selumetinib. Participants received selumetinib for a median of 41 cycles (min 2, max 67) at data cutoff. Approximately half of the children rated having some target tumor pain at baseline, which significantly decreased by pre-cycle 13. Most objectively measured baseline functions, including visual, motor, bowel/bladder, or airway function were within normal limits and did not clinically or statistically worsen during treatment. CONCLUSIONS Selumetinib resulted in PN shrinkage in most subjects with NF1 PN without clinically significant morbidity. No new PN-related symptoms developed while on selumetinib, and PRO measures indicated declines in tumor-related pain intensity. This supports that selumetinib treatment may prevent the development of PN-related morbidities, though future prospective studies are needed to confirm these results. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT01362803.
Collapse
Affiliation(s)
- Andrea M Gross
- Corresponding Authors: Andrea M. Gross, MD, NIH Clinical Center (Building 10), 10 Center Drive, Room 1-5742, Bethesda, MD 20852, USA ()
| | | | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Andrea Baldwin
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michael J Fisher
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - AeRang Kim
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC, USA
| | - Miriam Bornhorst
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC, USA
| | - Brian D Weiss
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Patricia Whitcomb
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Scott M Paul
- Rehabilitation Medicine Department, NIH Clinical Center, Baltimore, Maryland, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Baltimore, Maryland, USA
| | - David J Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Baltimore, Maryland, USA
| | - Staci Martin
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Amanda Carbonell
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kara Heisey
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Janet Therrien
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Oxana Kapustina
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Anne Dufek
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Joanne Derdak
- Pediatric Oncology Branch, Center for Cancer research, National Cancer Institute, Bethesda, Maryland, USA
| | - Malcolm A Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, USA (M.A.S.)
| | - Brigitte C Widemann
- Brigitte C. Widemann, MD, NIH Clinical Center (Building 10), 10 Center Drive, Room 1-3752, Bethesda, MD 20852, USA ()
| |
Collapse
|
36
|
Fink KR, Nobles SE, Zeitler DM. Comparing the Precision and Reliability Between Three Radiographic Techniques for Measuring Sporadic Vestibular Schwannomas. Acad Radiol 2022; 29:69-76. [PMID: 33158707 DOI: 10.1016/j.acra.2020.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 11/24/2022]
Abstract
RATIONALE AND OBJECTIVES Several methods exist for measuring vestibular schwannoma (VS) size radiographically. Our aim was to compare the precision and reproducibility of three different radiographic measurement techniques for assessing VS tumor size. MATERIAL AND METHODS Twenty patients with unilateral, sporadic VS previously untreated were identified. All patients had thin-slice T1 weighted, postcontrasted magnetization prepared rapid acquisition gradient echo images. Three measurement techniques were performed using within-subject and between-subject comparison. Experimental comparison of interobserver agreement between techniques was calculated. Interobserver intraclass correlation coefficients, repeatability coefficients, and relative smallest detectable difference were calculated and compared. RESULTS Mean tumor measurements were: 10.3 mm (maximum linear dimension, [MLD]), 495.9 mm3 (orthogonal volumetric analysis, [OVA]), and 572.1 mm3 (segmented volumetric analysis, [SVA]). Interobserver correlation coefficient was excellent for all measurement techniques, but highest for segmented volumetric analysis. Repeatability coefficient was 1.44 mm for MLD, 298.9 mm3 for OVA, and 174.8 mm3 for SVA. The smallest detectable difference was 13.9% for MLD, 60.2% for OVA, and 30.6% for SVA. A subgroup analysis was performed for small tumors (<14 mm) and large tumors (>14 mm) and demonstrated increased precision of segmented volumetric analysis for larger tumors. CONCLUSION Semi-automated segmented volumetric analysis appears more precise than either linear measurement or orthogonal volumetric analysis for reporting VS tumor size, and becomes increasingly precise for larger tumors. Tumor volume and tumor volume change over time using SVA may be more sensitive in surveilling VS than current measurement techniques.
Collapse
|
37
|
Sanchez LD, Bui A, Klesse LJ. Targeted Therapies for the Neurofibromatoses. Cancers (Basel) 2021; 13:cancers13236032. [PMID: 34885143 PMCID: PMC8657309 DOI: 10.3390/cancers13236032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Over the past several years, management of the tumors associated with the neurofibromatoses has been recognized to often require approaches that are distinct from their spontaneous counterparts. Focus has shifted to therapy aimed at minimizing symptoms given the risks of persistent, multiple tumors and new tumor growth. In this review, we will highlight the translation of preclinical data to therapeutic trials for patients with neurofibromatosis, particularly neurofibromatosis type 1 and neurofibromatosis type 2. Successful inhibition of MEK for patients with neurofibromatosis type 1 and progressive optic pathway gliomas or plexiform neurofibromas has been a significant advancement in patient care. Similar success for the malignant NF1 tumors, such as high-grade gliomas and malignant peripheral nerve sheath tumors, has not yet been achieved; nor has significant progress been made for patients with either neurofibromatosis type 2 or schwannomatosis, although efforts are ongoing.
Collapse
Affiliation(s)
- Lauren D. Sanchez
- Department of Pediatrics, Division of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Ashley Bui
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Laura J. Klesse
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX 75235, USA;
- Correspondence:
| |
Collapse
|
38
|
Welling DB, Collier KA, Burns SS, Oblinger JL, Shu E, Miles‐Markley BA, Hofmeister CC, Makary MS, Slone HW, Blakeley JO, Mansouri SA, Neff BA, Jackler RK, Mortazavi A, Chang L. Early phase clinical studies of AR-42, a histone deacetylase inhibitor, for neurofibromatosis type 2-associated vestibular schwannomas and meningiomas. Laryngoscope Investig Otolaryngol 2021; 6:1008-1019. [PMID: 34667843 PMCID: PMC8513424 DOI: 10.1002/lio2.643] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Two pilot studies of AR-42, a pan-histone deacetylase inhibitor, in human neurofibromatosis type 2 (NF2), vestibular schwannomas (VS), and meningiomas are presented. Primary endpoints included safety, and intra-tumoral pharmacokinetics (PK) and pharmacodynamics (PD). METHODS Pilot 1 is a subset analysis of a phase 1 study of AR-42 in solid tumors, which included NF2 or sporadic meningiomas. Tumor volumes and treatment-related adverse events (TRAEs) are reported (NCT01129193).Pilot 2 is a phase 0 surgical study of AR-42 assessing intra-tumoral PK and PD. AR-42 was administered for 3 weeks pre-operatively. Plasma and tumor drug concentrations and p-AKT expression were measured (NCT02282917). RESULTS Pilot 1: Five patients with NF2 and two with sporadic meningiomas experienced a similar incidence of TRAEs to the overall phase I trial. The six evaluable patients had 15 tumors (8 VS, 7 meningiomas). On AR-42, tumor volume increased in six, remained stable in eight, and decreased in one tumor. The annual percent growth rate decreased in eight, remained stable in three, and increased in four tumors. Pilot 2: Four patients with sporadic VS and one patient with meningioma experienced no grade 3/4 toxicities. Expression of p-AKT decreased in three of four VS. All tumors had higher AR-42 concentrations than plasma. CONCLUSIONS AR-42 is safe. Tumor volumes showed a mixed response, but most slowed growth. On a 40-mg regimen, drug concentrated in tumors and growth pathways were suppressed in most tumors, suggesting this may be a well-tolerated and effective dose. A phase 2 study of AR-42 for NF2-associated tumors appears warranted. LEVEL OF EVIDENCE 1b, 4.
Collapse
Affiliation(s)
- D. Bradley Welling
- Department of Otolaryngology Head and Neck SurgeryHarvard Medical School, Massachusetts Eye and Ear Infirmary, Massachusetts General HospitalBostonMassachusettsUSA
| | - Katharine A. Collier
- Division of Medical Oncology, Department of Internal MedicineThe Ohio State University College of Medicine and the Comprehensive Cancer CenterColumbusOhioUSA
| | - Sarah S. Burns
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Janet L. Oblinger
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Edina Shu
- Department of Otolaryngology Head and Neck SurgeryHarvard Medical School, Massachusetts Eye and Ear Infirmary, Massachusetts General HospitalBostonMassachusettsUSA
| | - Beth A. Miles‐Markley
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State University College of MedicineColumbusOhioUSA
| | - Craig C. Hofmeister
- Department of Hematology & OncologyWinship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Mina S. Makary
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - H. Wayne Slone
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - Jaishri O. Blakeley
- Departments of Neurology, Neurosurgery, & OncologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - S. Alireza Mansouri
- Departments of Neurology, Neurosurgery, & OncologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Brian A. Neff
- Department of Otolaryngology Head and Neck SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Robert K. Jackler
- Department of Otolaryngology Head and Neck SurgeryStanford UniversityPalo AltoCaliforniaUSA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal MedicineThe Ohio State University College of Medicine and the Comprehensive Cancer CenterColumbusOhioUSA
| | - Long‐Sheng Chang
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State University College of MedicineColumbusOhioUSA
| |
Collapse
|
39
|
Gross AM, Plotkin SR, Widemann BC. Neurofibromatosis Clinical Trials-REiNS Collaboration 2020 Recommendations: Looking Back and Moving Ahead. Neurology 2021; 97:S1-S3. [PMID: 34230201 PMCID: PMC11418092 DOI: 10.1212/wnl.0000000000012429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/19/2021] [Indexed: 11/15/2022] Open
Affiliation(s)
- Andrea M Gross
- From the Pediatric Oncology Branch (A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; and Department of Neurology and Cancer Center (S.R.P.), Massachusetts General Hospital, Boston.
| | - Scott R Plotkin
- From the Pediatric Oncology Branch (A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; and Department of Neurology and Cancer Center (S.R.P.), Massachusetts General Hospital, Boston
| | - Brigitte C Widemann
- From the Pediatric Oncology Branch (A.M.G., B.C.W.), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD; and Department of Neurology and Cancer Center (S.R.P.), Massachusetts General Hospital, Boston
| |
Collapse
|
40
|
Casey D, Demko S, Sinha A, Mishra-Kalyani PS, Shen YL, Khasar S, Goheer MA, Helms WS, Pan L, Xu Y, Fan J, Leong R, Liu J, Yang Y, Windsor K, Ou M, Stephens O, Oh B, Reaman GH, Nair A, Shord SS, Bhatnagar V, Daniels SR, Sickafuse S, Goldberg KB, Theoret MR, Pazdur R, Singh H. FDA Approval Summary: Selumetinib for Plexiform Neurofibroma. Clin Cancer Res 2021; 27:4142-4146. [PMID: 33712511 DOI: 10.1158/1078-0432.ccr-20-5032] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 11/16/2022]
Abstract
On April 10, 2020, the FDA approved selumetinib (KOSELUGO, AstraZeneca) for the treatment of pediatric patients 2 years of age and older with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas. Approval was based on demonstration of a durable overall response rate per Response Evaluation in Neurofibromatosis and Schwannomatosis criteria and supported by observed clinical improvements in plexiform neurofibroma-related symptoms and functional impairments in 50 pediatric patients with inoperable plexiform neurofibromas in a single-arm, multicenter trial. The overall reponse rate per NCI investigator assessment was 66% (95% confidence interval, 51-79) with at least 12 months of follow-up. The median duration of response was not reached, and 82% of responding patients experienced duration of response ≥12 months. Clinical outcome assessment endpoints provided supportive efficacy data. Risks of selumetinib are consistent with MAPK (MEK) inhibitor class effects, including ocular, cardiac, musculoskeletal, gastrointestinal, and dermatologic toxicities. Safety was assessed across a pooled database of 74 pediatric patients with plexiform neurofibromas and supported by adult and pediatric selumetinib clinical trial data in cancer indications. The benefit-risk assessment for selumetinib in patients with inoperable plexiform neurofibromas was considered favorable.
Collapse
Affiliation(s)
- Denise Casey
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | - Suzanne Demko
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | - Arup Sinha
- Office of Biostatistics, FDA, Silver Spring, Maryland
| | | | - Yuan-Li Shen
- Office of Biostatistics, FDA, Silver Spring, Maryland
| | - Sachia Khasar
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | - M Anwar Goheer
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland
| | | | - Lili Pan
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Yuan Xu
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Jianghong Fan
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Ruby Leong
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Jiang Liu
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | - Yuching Yang
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | | | - Mei Ou
- Office of Pharmaceutical Quality, FDA, Silver Spring, Maryland
| | - Olen Stephens
- Office of Pharmaceutical Quality, FDA, Silver Spring, Maryland
| | - Byeongtaek Oh
- Office of Pharmaceutical Quality, FDA, Silver Spring, Maryland
| | | | - Abhilasha Nair
- Oncology Center of Excellence, FDA, Silver Spring, Maryland
| | - Stacy S Shord
- Office of Clinical Pharmacology, FDA, Silver Spring, Maryland
| | | | - Selena R Daniels
- Division of Clinical Outcome Assessment, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland
| | | | | | - Marc R Theoret
- Oncology Center of Excellence, FDA, Silver Spring, Maryland
| | - Richard Pazdur
- Oncology Center of Excellence, FDA, Silver Spring, Maryland
| | - Harpreet Singh
- Office of Oncologic Diseases, FDA, Silver Spring, Maryland. .,Oncology Center of Excellence, FDA, Silver Spring, Maryland
| |
Collapse
|
41
|
Assessment of Tumor Volume Dynamics and Outcome After Radiosurgery for the Treatment of Vestibular Schwannoma: A Single-Center Experience. Otol Neurotol 2021; 42:e750-e757. [PMID: 34111052 DOI: 10.1097/mao.0000000000003056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To assess the factors affecting early local and audiometric outcomes in vestibular schwannoma (VS) patients treated with stereotactic radiosurgery (SRS). STUDY DESIGN A retrospective review of medical records. SETTING Tertiary referral center. PATIENTS Records of all adult patients who underwent SRS between 2010 and 2016 for the treatment of VS were retrospectively reviewed. Patients treated with microsurgery or multi-fractionation schemes, and those who had neurofibromatosis type 2, were excluded. INTERVENTION SRS, tumor volume/size measurements. MAIN OUTCOME MEASURES The impact of tumor volume dynamics on the early local and hearing-related outcomes, together with the factors that influence them following SRS, and comparison of different tumor size measurement methods. RESULTS From 2010 to 2016, 53 patients underwent single fraction SRS of 12 Gy. Median follow-up time was 32 months (range, 6-79). At the last follow-up, only one patient had clinical progression. Age less than or equal to 65 years (p = 0.04; odds ratio [OR]: 0.17; 95% confidence interval [CI]: 0.03-0.93) and baseline pure-tone average (PTA) level less than or equal to 30 dB (p = 0.03; OR: 0.90; 95% CI: 0.84-0.96) were associated with maintenance of serviceable hearing. On multivariate analysis, PTA remained significant (p = 0.01; OR: 0.04; 95% CI: 0.003-0.45). In patients with a loss of serviceable hearing, the mean volume increase tended to be higher than in the patients whose hearing was maintained. The linear measurement method underestimated, and the A × B × C/2 equation overestimated, the radiological progression compared with 3D-volumetric delineations. CONCLUSION During the median observation period of almost 3 years, we reported our early outcome results. Tumor volume increase may have an impact on serviceable hearing loss after SRS. Currently there is no widely accepted method for the evaluation of post-SRS response. Linear measurement and the A × B × C/2 equation produce less reliable estimates of radiological progression compared with 3D-volumetric delineations. Accurate volume measurements with 3D delineations should be considered as part of clinical routine for assessing progression and deciding on salvage therapies.
Collapse
|
42
|
Thalheimer RD, Merker VL, Ly KI, Champlain A, Sawaya J, Askenazi NL, Herr HP, Da JLW, Jordan JT, Muzikansky A, Pearce EM, Sakamoto FH, Blakeley JO, Anderson RR, Plotkin SR. Validating Techniques for Measurement of Cutaneous Neurofibromas: Recommendations for Clinical Trials. Neurology 2021; 97:S32-S41. [PMID: 34230197 DOI: 10.1212/wnl.0000000000012428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/11/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the reliability and variability of digital calipers, 3D photography, and high-frequency ultrasound (HFUS) for measurement of cutaneous neurofibromas (cNF) in patients with neurofibromatosis type 1 (NF1). BACKGROUND cNF affect virtually all patients with NF1 and are a major source of morbidity. Reliable techniques for measuring cNF are needed to develop therapies for these tumors. METHODS Adults with NF1 were recruited. For each participant, 6 cNF were assessed independently by 3 different examiners at 5 different time points using digital calipers, 3D photography, and HFUS. The intraclass correlation coefficient (ICC) was used to assess intrarater and interrater reliability of linear and volumetric measurements for each technique, with ICC values >0.90 defined as excellent reliability. The coefficient of variation (CV) was used to estimate the minimal detectable difference (MDD) for each technique. RESULTS Fifty-seven cNF across 10 participants were evaluated. The ICC for image acquisition and measurement was >0.97 within and across examiners for HFUS and 3D photography. ICC for digital calipers was 0.62-0.88. CV varied by measurement tool, linear vs volumetric measurement, and tumor size. CONCLUSIONS HFUS and 3D photography demonstrate excellent reliability whereas digital calipers have good to excellent reliability in measuring cNF. The MDD for each technique was used to create tables of proposed thresholds for investigators to use as guides for clinical trials focused on cNF size. These criteria should be updated as the performance of these end points is evaluated.
Collapse
Affiliation(s)
- Raquel D Thalheimer
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vanessa L Merker
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - K Ina Ly
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Amanda Champlain
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jennifer Sawaya
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Naomi L Askenazi
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hamilton P Herr
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jennifer L W Da
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Justin T Jordan
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alona Muzikansky
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth Morehouse Pearce
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Fernanda H Sakamoto
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jaishri O Blakeley
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - R Rox Anderson
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Scott R Plotkin
- From the Department of Neurology and Cancer Center (R.T., V.L.M., I.L., N.L.A., H.P.H., J.L.W.D., J.T.J., S.R.P.), Wellman Center for Photomedicine (A.C., J.S., E.M.P., F.H.S., R.R.A.), and Biostatistics Center (A.M.), Massachusetts General Hospital, and Department of Dermatology (A.C., J.S., E.M.P., F.H.S., R.R.A.), Harvard Medical School, Boston; and Department of Neurology, Neurosurgery, and Oncology (J.B.), Johns Hopkins University School of Medicine, Baltimore, MD.
| | | |
Collapse
|
43
|
Gross AM. Using real world data to support regulatory approval of drugs in rare diseases: A review of opportunities, limitations & a case example. Curr Probl Cancer 2021; 45:100769. [PMID: 34247834 DOI: 10.1016/j.currproblcancer.2021.100769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/21/2021] [Indexed: 11/27/2022]
Abstract
Conducting clinical research in patients with rare diseases presents a variety of challenges. At the same time, rare diseases represent a large area of unmet medical need with a significant burden of morbidity throughout the world. One of the most common issues with designing clinical trials for rare disease populations is that the gold-standard randomized controlled trial design is often not feasible in these small and usually geographically dispersed populations. Real world data therefore has particular relevance in the rare disease setting, where it may be used as a comparator for single-arm treatment trials and in support of submissions to regulatory agencies for drugs to treat these conditions. In this report, we review the potential utility and limitations of external controls for regulatory approval of drugs in rare diseases and present a recent case example of the successful utilization of external controls in the Neurofibromatosis type 1 (NF1) population.
Collapse
|
44
|
US Food and Drug Administration regulatory updates in neuro-oncology. J Neurooncol 2021; 153:375-381. [PMID: 34156585 PMCID: PMC8218275 DOI: 10.1007/s11060-021-03789-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/11/2021] [Indexed: 11/21/2022]
Abstract
Objective Contemporary management of patients with neuro-oncologic disease requires an understanding of approvals by the US Food and Drug Administration (FDA) related to nervous system tumors. To summarize FDA updates applicable to neuro-oncology practitioners, we sought to review oncology product approvals and Guidances that were pertinent to the field in the past year. Methods Oncology product approvals between January 1, 2020, and December 31, 2020, were reviewed for clinical trial outcomes involving tumors of the nervous system. FDA Guidances relevant to neuro-oncology were also reviewed. Results Five oncology product approvals described outcomes for nervous system tumors in the year 2020. These included the first regulatory approval for neurofibromatosis type 1: selumetinib for children with symptomatic, inoperable plexiform neurofibromas. Additionally, there were 4 regulatory approvals for non-central nervous system (CNS) cancers that described clinical outcomes for patients with brain metastases. These included the approval of tucatinib for metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer including patients with brain metastases, brigatinib for anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC), and pralsetinib and selpercatinib for RET fusion-positive NSCLC. Finally, two FDA Guidances for Industry, “Cancer Clinical Trial Eligibility Criteria: Brain Metastases” and “Evaluating Cancer Drugs in Patients with Central Nervous System Metastases” were published to facilitate drug development for and inclusion of patients with CNS metastases in clinical trials. Conclusions Despite the challenges of the past year brought on by the COVID-19 pandemic, progress continues to be made in neuro-oncology. These include first-of-their-kind FDA approvals and Guidances that are relevant to the management of patients with nervous system tumors.
Collapse
|
45
|
Ronsley R, Hounjet CD, Cheng S, Rassekh SR, Duncan WJ, Dunham C, Gardiner J, Ghag A, Ludemann JP, Wensley D, Rehmus W, Sargent MA, Hukin J. Trametinib therapy for children with neurofibromatosis type 1 and life-threatening plexiform neurofibroma or treatment-refractory low-grade glioma. Cancer Med 2021; 10:3556-3564. [PMID: 33939292 PMCID: PMC8178485 DOI: 10.1002/cam4.3910] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose To describe a series of children with extensive PNF or treatment refractory PLGG treated on a compassionate basis with trametinib. Methods We report on six patients with NF‐1 treated with trametinib on a compassionate basis at British Columbia Children's Hospital since 2017. Data were collected retrospectively from the patient record. RAPNO and volumetric criteria were used to evaluate the response of intracranial and extracranial lesions, respectively. Results Subjects were 21 months to 14 years old at the time of initiation of trametinib therapy and 3/6 subjects are male. Duration of therapy was 4–28 months at the time of this report. All patients had partial response or were stable on analysis. Two patients with life‐threatening PNF had a partial radiographic response in tandem with significant clinical improvement and developmental catch up. One subject discontinued therapy after 6 months due to paronychia and inadequate response. The most common adverse effect (AE) was grade 1–2 paronychia or dermatitis in 5/6 patients. There were no grade 3 or 4 AEs. At the time of this report, five patients remain on therapy. Conclusion Trametinib is an effective therapy for advanced PNF and refractory PLGG in patients with NF‐1 and is well tolerated in children. Further data and clinical trials are required to assess tolerance, efficacy and durability of response, and length of treatment required in such patients.
Collapse
Affiliation(s)
- Rebecca Ronsley
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Celine D Hounjet
- Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Sylvia Cheng
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Shahrad Rod Rassekh
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Walter J Duncan
- Division of Pediatric Cardiology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Christopher Dunham
- Division of Anatomic Pathology, Department of Pathology, University of British Columbia, Vancouver, Canada
| | - Jane Gardiner
- Division of Pediatric Ophthalmology, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Arvindera Ghag
- Division of Pediatric Orthopedic Surgery, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - Jeffrey P Ludemann
- Division of Pediatric Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, Canada
| | - David Wensley
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Wingfield Rehmus
- Division of Dermatology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Michael A Sargent
- Division of Pediatric Neuro-Radiology, Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Juliette Hukin
- Division of Hematology, Oncology & BMT, Department of Pediatrics, University of British Columbia, Vancouver, Canada.,Division of Neurology, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
46
|
Ardern-Holmes S, White C, Bahure S, So S, McCowage G, Hovey E, Troon S, De Souza P, Simes J, Slancar M, Dexter M, Wong M. The Effect of Bevacizumab on Vestibular Schwannoma Related to Neurofibromatosis Type 2. AUSTRALASIAN JOURNAL OF NEUROSCIENCE 2021. [DOI: 10.21307/ajon-2021-002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
47
|
Population pharmacokinetics and exposure-response of selumetinib and its N-desmethyl metabolite in pediatric patients with neurofibromatosis type 1 and inoperable plexiform neurofibromas. Cancer Chemother Pharmacol 2021; 88:189-202. [PMID: 33903938 DOI: 10.1007/s00280-021-04274-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Selumetinib (ARRY-142886) is a potent, selective, MEK1/2 inhibitor approved in the US for the treatment of children (≥ 2 years) with neurofibromatosis type 1 (NF1) and symptomatic, inoperable plexiform neurofibromas (PN). We characterized population pharmacokinetics (PK) of selumetinib and its active N-desmethyl metabolite, evaluated exposure-safety/efficacy relationships, and assessed the proposed therapeutic dose of 25 mg/m2 bid based on body surface area (BSA) in this patient population. METHODS Population PK modeling and covariate analysis (demographics, formulation, liver enzymes, BSA, patients/healthy volunteers) were based on pooled PK data from adult healthy volunteers (n = 391), adult oncology patients (n = 83) and pediatric patients with NF1-PN (n = 68). Longitudinal selumetinib/metabolite exposures were predicted with the final model. Exposure-safety/efficacy analyses were applied to pediatric patients (dose levels: 20, 25, 30 mg/m2 bid). RESULTS Selumetinib and metabolite concentration-time courses were modeled using a joint compartmental model. Typical selumetinib plasma clearance was 11.6 L/h (95% CI 11.0-12.2 L/ h). Only BSA had a clinically relevant (> 20%) impact on exposure, supporting BSA-based administration in children. Selumetinib and metabolite exposures in responders (≥ 20% PN volume decrease from baseline) and non-responders were largely overlapping, with medians numerically higher in responders. No clear relationships between exposure and safety events were established; exposure was not associated with key adverse events (AEs) including rash acneiform, diarrhea, vomiting, and nausea. CONCLUSION Findings support continuous selumetinib 25 mg/m2 bid in pediatric patients. Importantly, the updated dosing nomogram ensures that patients will receive a clinically active, yet tolerable, dose regardless of differences in BSA and allows dose reductions, if necessary.
Collapse
|
48
|
Harder A. MEK inhibitors - novel targeted therapies of neurofibromatosis associated benign and malignant lesions. Biomark Res 2021; 9:26. [PMID: 33863389 PMCID: PMC8052700 DOI: 10.1186/s40364-021-00281-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
MAP/ERK kinase 1 and 2 (MEK 1/2) inhibitors (MEKi) are investigated in several trials to treat lesions that arise from pathogenic variants of the Neurofibromatosis type 1 and type 2 genes (NF1, NF2). These trials showed that MEKi are capable to shrink volume of low grade gliomas and plexiform neurofibromas in NF1. Targeting other lesions being associated with a high morbidity in NF1 seems to be promising. Due to involvement of multiple pathways in NF2 associated lesions as well as in malignant tumors, MEKi are also used in combination therapies. This review outlines the current state of MEKi application in neurofibromatosis and associated benign and malignant lesions.
Collapse
Affiliation(s)
- Anja Harder
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06120, Halle (Saale), Germany. .,Institute of Neuropathology, University Hospital Münster, Münster, Germany. .,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany.
| |
Collapse
|
49
|
Shi C, Zhou Z, Lin H, Gao J. Imaging Beyond Seeing: Early Prognosis of Cancer Treatment. SMALL METHODS 2021; 5:e2001025. [PMID: 34927817 DOI: 10.1002/smtd.202001025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Indexed: 06/14/2023]
Abstract
Assessing cancer response to therapeutic interventions has been realized as an important course to early predict curative efficacy and treatment outcomes due to tumor heterogeneity. Compared to the traditional invasive tissue biopsy method, molecular imaging techniques have fundamentally revolutionized the ability to evaluate cancer response in a spatiotemporal manner. The past few years has witnessed a paradigm shift on the efforts from manufacturing functional molecular imaging probes for seeing a tumor to a vantage stage of interpreting the tumor response during different treatments. This review is to stand by the current development of advanced imaging technologies aiming to predict the treatment response in cancer therapy. Special interest is placed on the systems that are able to provide rapid and noninvasive assessment of pharmacokinetic drug fates (e.g., drug distribution, release, and activation) and tumor microenvironment heterogeneity (e.g., tumor cells, macrophages, dendritic cells (DCs), T cells, and inflammatory cells). The current status, practical significance, and future challenges of the emerging artificial intelligence (AI) technology and machine learning in the applications of medical imaging fields is overviewed. Ultimately, the authors hope that this review is timely to spur research interest in molecular imaging and precision medicine.
Collapse
Affiliation(s)
- Changrong Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zijian Zhou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hongyu Lin
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jinhao Gao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
50
|
Osum SH, Watson AL, Largaespada DA. Spontaneous and Engineered Large Animal Models of Neurofibromatosis Type 1. Int J Mol Sci 2021; 22:1954. [PMID: 33669386 PMCID: PMC7920315 DOI: 10.3390/ijms22041954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Animal models are crucial to understanding human disease biology and developing new therapies. By far the most common animal used to investigate prevailing questions about human disease is the mouse. Mouse models are powerful tools for research as their small size, limited lifespan, and defined genetic background allow researchers to easily manipulate their genome and maintain large numbers of animals in general laboratory spaces. However, it is precisely these attributes that make them so different from humans and explains, in part, why these models do not accurately predict drug responses in human patients. This is particularly true of the neurofibromatoses (NFs), a group of genetic diseases that predispose individuals to tumors of the nervous system, the most common of which is Neurofibromatosis type 1 (NF1). Despite years of research, there are still many unanswered questions and few effective treatments for NF1. Genetically engineered mice have drastically improved our understanding of many aspects of NF1, but they do not exemplify the overall complexity of the disease and some findings do not translate well to humans due to differences in body size and physiology. Moreover, NF1 mouse models are heavily reliant on the Cre-Lox system, which does not accurately reflect the molecular mechanism of spontaneous loss of heterozygosity that accompanies human tumor development. Spontaneous and genetically engineered large animal models may provide a valuable supplement to rodent studies for NF1. Naturally occurring comparative models of disease are an attractive prospect because they occur on heterogeneous genetic backgrounds and are due to spontaneous rather than engineered mutations. The use of animals with naturally occurring disease has been effective for studying osteosarcoma, lymphoma, and diabetes. Spontaneous NF-like symptoms including neurofibromas and malignant peripheral nerve sheath tumors (MPNST) have been documented in several large animal species and share biological and clinical similarities with human NF1. These animals could provide additional insight into the complex biology of NF1 and potentially provide a platform for pre-clinical trials. Additionally, genetically engineered porcine models of NF1 have recently been developed and display a variety of clinical features similar to those seen in NF1 patients. Their large size and relatively long lifespan allow for longitudinal imaging studies and evaluation of innovative surgical techniques using human equipment. Greater genetic, anatomic, and physiologic similarities to humans enable the engineering of precise disease alleles found in human patients and make them ideal for preclinical pharmacokinetic and pharmacodynamic studies of small molecule, cellular, and gene therapies prior to clinical trials in patients. Comparative genomic studies between humans and animals with naturally occurring disease, as well as preclinical studies in large animal disease models, may help identify new targets for therapeutic intervention and expedite the translation of new therapies. In this review, we discuss new genetically engineered large animal models of NF1 and cases of spontaneous NF-like manifestations in large animals, with a special emphasis on how these comparative models could act as a crucial translational intermediary between specialized murine models and NF1 patients.
Collapse
Affiliation(s)
- Sara H. Osum
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - David A. Largaespada
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|