1
|
Lee V, Hultcrantz M, Petrone S, Lewis EW, Banna H, Lichtman E, Thulasi P, Quick AA, Jeng BH, Sunshine SB, Francis JH, Canestraro J, Farooq AV, Clements P, Robertson N, Burman M, McKevitt T, Struemper H, Weir L. Characterization of Belantamab Mafodotin-Induced Corneal Changes in Patients With Multiple Myeloma. JAMA Ophthalmol 2025:2833594. [PMID: 40338596 PMCID: PMC12062993 DOI: 10.1001/jamaophthalmol.2025.1008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/09/2025] [Indexed: 05/09/2025]
Abstract
Importance Ocular surface events are a class effect of microtubule-inhibitor payload-containing antibody-drug conjugates (ADCs); the mechanism underlying these events has not been fully elucidated. Objective To characterize corneal epithelial changes in patients with relapsed or refractory multiple myeloma (RRMM) treated with belantamab mafodotin, a maleimidocaproyl monomethyl auristatin-F (MMAF)-containing ADC. Design, Setting, and Participants This multicenter, phase 3b case series study was conducted in the US from March 26, 2020, to November 21, 2022, among adults with RRMM. Data were analyzed from May 2021 to May 2023. Exposure Prior or ongoing treatment with belantamab mafodotin. Main Outcomes and Measures The primary end point included pathologic characteristics and composition of corneal epithelial changes obtained by impression cytology (IC) or superficial keratectomy (SK) in patients treated with belantamab mafodotin. Tear film and blood were collected to determine belantamab mafodotin concentrations in patients at the time of sampling. Results Of 16 patients screened, 9 were included in this study, with 6 evaluable corneal samples obtained from 6 patients either by IC (n = 4) or SK (n = 2). Of 9 patients included, median (range) patient age was 67.0 (57.0-81.0) years for those with samples obtained by IC and 68.0 (65.0-81.0) years for those with samples obtained by SK. Six patients (67%) were female. All samples demonstrated epithelial cells with eosinophilic intracytoplasmic inclusions, basophilic granular cytoplasm, or both. Five samples were positive for apoptosis, and 3 samples showed evidence of inflammation. All patients experienced complete IC or SK wound healing. ADC was detected in the tear fluid of 5 of 7 patients with tear fluid sampling, while ADC was quantifiable in 3 of 4 patients with blood samples. Conclusions and Relevance In this case series study, intracytoplasmic inclusions were observed by histopathology in the corneal epithelium of patients exposed to belantamab mafodotin, and the pattern of corneal changes suggests limbal vessels may be a primary pathway enabling ADC to reach the cornea. Although limited to 6 samples, this study helps us better understand corneal changes associated with certain ADCs. Trial Registration ClinicalTrials.gov Identifier: NCT04549363.
Collapse
Affiliation(s)
- Vivian Lee
- Department of Ophthalmology and Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Malin Hultcrantz
- Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Hussam Banna
- Department of Ophthalmology, University of North Carolina at Chapel Hill
| | - Eben Lichtman
- Division of Hematology, University of North Carolina at Chapel Hill
| | - Praneetha Thulasi
- Emory University, Atlanta, Georgia
- Now with Ophthalmology and Visual Sciences Division of Cornea and External Disease, Washington University Physicians, St Louis, Missouri
| | - Anjulie A. Quick
- Department of Ophthalmology, University of Kansas Medical Center, Prairie Village
| | - Bennie H. Jeng
- Ophthalmology and Visual Sciences, University of Maryland, Baltimore
- Now with Department of Ophthalmology and Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Sarah B. Sunshine
- Ophthalmology and Visual Sciences, University of Maryland, Baltimore
| | - Jasmine H. Francis
- Ophthalmic Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julia Canestraro
- Ophthalmic Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Asim V. Farooq
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Peter Clements
- Pathology, Nonclinical Safety, GSK, Stevenage, United Kingdom
| | | | - Mark Burman
- Formerly with Department of Genetic Toxicology, GSK, Ware, United Kingdom
| | - Tom McKevitt
- Pathology, Nonclinical Safety, GSK, Stevenage, United Kingdom
- Now with Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, United Kingdom
| | - Herbert Struemper
- Clinical Pharmacology Modeling & Simulation, GSK, Durham, North Carolina
| | - Lucinda Weir
- Nonclinical Safety, GSK, Stevenage, United Kingdom
| |
Collapse
|
2
|
Tripathy DK, Panda LP, Biswal S, Barhwal K. Insights into the glioblastoma tumor microenvironment: current and emerging therapeutic approaches. Front Pharmacol 2024; 15:1355242. [PMID: 38523646 PMCID: PMC10957596 DOI: 10.3389/fphar.2024.1355242] [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: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 03/26/2024] Open
Abstract
Glioblastoma (GB) is an intrusive and recurrent primary brain tumor with low survivability. The heterogeneity of the tumor microenvironment plays a crucial role in the stemness and proliferation of GB. The tumor microenvironment induces tumor heterogeneity of cancer cells by facilitating clonal evolution and promoting multidrug resistance, leading to cancer cell progression and metastasis. It also plays an important role in angiogenesis to nourish the hypoxic tumor environment. There is a strong interaction of neoplastic cells with their surrounding microenvironment that comprise several immune and non-immune cellular components. The tumor microenvironment is a complex network of immune components like microglia, macrophages, T cells, B cells, natural killer (NK) cells, dendritic cells and myeloid-derived suppressor cells, and non-immune components such as extracellular matrix, endothelial cells, astrocytes and neurons. The prognosis of GB is thus challenging, making it a difficult target for therapeutic interventions. The current therapeutic approaches target these regulators of tumor micro-environment through both generalized and personalized approaches. The review provides a summary of important milestones in GB research, factors regulating tumor microenvironment and promoting angiogenesis and potential therapeutic agents widely used for the treatment of GB patients.
Collapse
Affiliation(s)
- Dev Kumar Tripathy
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Lakshmi Priya Panda
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Kalpana Barhwal
- Department of Physiology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| |
Collapse
|
3
|
Suzuki Y, Zhou S, Ota Y, Harrington M, Miyagi E, Takagi H, Kuno T, Wright JD. Toxicity profiles of antibody-drug conjugates for anticancer treatment: a systematic review and meta-analysis. JNCI Cancer Spectr 2023; 7:pkad069. [PMID: 37756687 PMCID: PMC10579782 DOI: 10.1093/jncics/pkad069] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/06/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Antibody-drug conjugates are attractive targeted agents in anticancer treatment because of their unique mechanism of action and reduced toxicity. Little is known about the spectrum of adverse events associated with antibody-drug conjugates, despite tens of clinical trials. METHODS A systematic review of randomized controlled trials evaluating antibody-drug conjugate efficacy in anticancer treatment was conducted. PubMed, EMBASE, and ClinicalTrial.gov were searched for relevant studies. Meta-analyses assessed the odds ratios (ORs) of 12 treatment-related symptoms and toxicities in patients treated with antibody-drug conjugates compared with those receiving other anticancer agents without antibody-drug conjugates. All-grade and high-grade (grade ≥3) toxicities were examined. RESULTS Twenty studies involving 10 075 patients were included. Compared with control groups, antibody-drug conjugates were associated with a higher risk of all-grade fatigue (OR = 1.25, 95% confidence interval [CI] = 1.08 to 1.45), anorexia (OR = 1.36, 95% CI = 1.09 to 1.69), nausea (OR = 1.46, 95% CI = 1.09 to 1.97), and sensory neuropathy (OR = 2.18, 95% CI = 1.27 to 3.76) as treatment-related symptoms. Patients treated with antibody-drug conjugates had a statistically significantly lower risk of all-grade febrile neutropenia (OR = 0.46, 95% CI = 0.22 to 0.96). Conversely, they had a higher risk of thrombocytopenia (OR = 2.07, 95% CI = 1.00 to 4.31), increased alanine aminotransferase (OR = 2.51, 95% CI = 1.84 to 3.40), and increased aspartate aminotransferase (OR = 2.83, 95% CI = 2.04 to 3.93). Subgroup analysis showed a similar toxicity profile when comparing the solid tumors with hematologic malignancy groups and the antibody-drug conjugate vs antibody-drug conjugate plus chemotherapy groups, except for some neurologic and hematologic adverse events. CONCLUSIONS This comprehensive profile of adverse events associated with antibody-drug conjugate-based treatment shows an increase in various types of all-grade treatment-related symptoms and adverse events, although no increase in high-grade adverse events was seen.
Collapse
Affiliation(s)
- Yukio Suzuki
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY, USA
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Susu Zhou
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, New York, NY, USA
| | - Yukihide Ota
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew Harrington
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, New York, NY, USA
| | - Etsuko Miyagi
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hisato Takagi
- Department of Cardiovascular Surgery, Shizuoka Medical Center, Shizuoka, Japan
| | - Toshiki Kuno
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY, USA
| | - Jason D Wright
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
4
|
Gan HK, Parakh S, Osellame LD, Cher L, Uccellini A, Hafeez U, Menon S, Scott AM. Antibody drug conjugates for glioblastoma: current progress towards clinical use. Expert Opin Biol Ther 2023; 23:1089-1102. [PMID: 37955063 DOI: 10.1080/14712598.2023.2282729] [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: 08/20/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Antibody drug conjugates (ADCs) are now a proven therapeutic class for many cancers, combining highly specific targeting with the potency of high effective payloads. This review summarizes the experience with ADCs in brain tumors and examines future paths for their use in these tumors. AREAS COVERED This review will cover all the key classes of ADCs which have been tested in primary brain tumors, including commentary on the major trials to date. The efficacy of these trials, as well as their limitations, will put in context of the overall landscape of drug development in brain tumors. Importantly, this review will summarize key learnings and insights from these trials that help provide the basis for rational ways in which these drugs can be effectively and appropriate developed for patients with primary brain tumors. EXPERT OPINION ADC development in brain tumors has occurred in two major phases to date. Key learnings from previous trials provide a strong rationale for the continued development of these drugs for primary brain tumors. However, the unique biology of these tumors requires development strategies specifically tailored to maximize their optimal development.
Collapse
Affiliation(s)
- Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Sagun Parakh
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Laura D Osellame
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Biochemistry and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Heidelberg, Melbourne, Victoria, Australia
| | - Lawrence Cher
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
| | | | - Umbreen Hafeez
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
| | - Siddharth Menon
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Andrew M Scott
- Tumour Targeting Program, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
5
|
Vize CJ, Kim SK, Matthews T, Macsai M, Merrell R, Hsu S, Kundu MG, Yoon J, Kennedy E, Pai M, Bain E, Lassman AB, Moazami G. A Phase 3b Study for Management of Ocular Side Effects in Patients with Epidermal Growth Factor Receptor-Amplified Glioblastoma Receiving Depatuxizumab Mafodotin. Ophthalmic Res 2023; 66:1030-1043. [PMID: 37257422 PMCID: PMC10413800 DOI: 10.1159/000531142] [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: 10/14/2021] [Accepted: 05/09/2023] [Indexed: 06/02/2023]
Abstract
INTRODUCTION The Understanding New Interventions with GBM ThErapy (UNITE) study was designed to assess the effect of prophylaxis for ocular side effects (OSEs) in patients with glioblastoma receiving the antibody-drug conjugate (ADC) depatuxizumab mafodotin. UNITE (NCT03419403) was a phase 3b, open-label, randomized, exploratory study performed at 18 research sites in 5 countries. METHODS The study enrolled adult patients with epidermal growth factor receptor-amplified, histologically confirmed, newly diagnosed supratentorial glioblastoma or grade IV gliosarcoma, and a Karnofsky Performance Status ≥70, receiving depatuxizumab mafodotin. All patients were administered depatuxizumab mafodotin during concurrent radiotherapy and temozolomide and with adjuvant temozolomide. Ninety patients were to be randomized (1:1:1) to OSE prophylactic treatments with each depatuxizumab mafodotin infusion: (a) standard steroid eye drops, (b) standard steroid eye drops plus vasoconstrictor eye drops and cold compress, or (c) enhanced steroids plus vasoconstrictor eye drops and cold compress. A Corneal Epitheliopathy Adverse Event (CEAE) scale was devised to capture symptoms, grade OSEs (scale of 0-5), and inform ADC dose modifications. The primary endpoint was the frequency of a required change in OSE management due to inadequate control of OSEs, defined as decline from baseline in visual acuity (using logarithm of the minimum angle of resolution [LogMAR] scale) or a Grade ≥3 CEAE event, in the worst eye in the first 8 weeks of treatment; unless otherwise specified, the treatment period refers to both the chemoradiation and adjuvant phases. RESULTS The UNITE study was stopped early after interim analysis of separate phase III trial showed no difference in survival from depatuxizumab mafodotin. Forty patients were randomized (38 received depatuxizumab mafodotin). Overall, 23 patients experienced inadequate control of OSEs that required change in OSE management within 8 weeks of treatment, with 21 (70.0%) experiencing ≥+0.3 change on LogMAR scale in baseline-adjusted visual acuity and 12 reporting a grade ≥3 CEAE. There were no definitive differences among prophylactic treatments. CONCLUSIONS The premature cessation of the study precludes definitive conclusions regarding the OSE prophylaxis strategies. No new clinically significant safety findings were noted. Despite these limitations, this study highlights the need for novel assessment tools to better understand and mitigate OSEs associated with ADCs.
Collapse
Affiliation(s)
- Colin J. Vize
- Department of Ophthalmology, Hull University Teaching Hospitals NHS Trust, Hull, UK
| | - Stella K. Kim
- Department of Ophthalmology and Visual Science, University of Texas McGovern Medical School, Houston, TX, USA
| | - Tim Matthews
- Birmingham Neuro-Ophthalmology Unit, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Marian Macsai
- Northshore University Health System, Glenview, IL, USA
| | - Ryan Merrell
- NorthShore University Health System, Evanston, IL, USA
| | - Sigmund Hsu
- The Vivian L. Smith Department of Neurosurgery, University of Texas McGovern Medical School, Houston, TX, USA
| | | | | | | | | | | | - Andrew B. Lassman
- Division of Neuro-Oncology, Department of Neurology and the Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian, New York, NY, USA
| | - Golnaz Moazami
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
6
|
Mair MJ, Bartsch R, Le Rhun E, Berghoff AS, Brastianos PK, Cortes J, Gan HK, Lin NU, Lassman AB, Wen PY, Weller M, van den Bent M, Preusser M. Understanding the activity of antibody-drug conjugates in primary and secondary brain tumours. Nat Rev Clin Oncol 2023; 20:372-389. [PMID: 37085569 DOI: 10.1038/s41571-023-00756-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/23/2023]
Abstract
Antibody-drug conjugates (ADCs), a class of targeted cancer therapeutics combining monoclonal antibodies with a cytotoxic payload via a chemical linker, have already been approved for the treatment of several cancer types, with extensive clinical development of novel constructs ongoing. Primary and secondary brain tumours are associated with high mortality and morbidity, necessitating novel treatment approaches. Pharmacotherapy of brain tumours can be limited by restricted drug delivery across the blood-brain or blood-tumour barrier, although data from phase II studies of the HER2-targeted ADC trastuzumab deruxtecan indicate clinically relevant intracranial activity in patients with brain metastases from HER2+ breast cancer. However, depatuxizumab mafodotin, an ADC targeting wild-type EGFR and EGFR variant III, did not provide a definitive overall survival benefit in patients with newly diagnosed or recurrent EGFR-amplified glioblastoma in phase II and III trials, despite objective radiological responses in some patients. In this Review, we summarize the available data on the central nervous system activity of ADCs from trials involving patients with primary and secondary brain tumours and discuss their clinical implications. Furthermore, we explore pharmacological determinants of intracranial activity and discuss the optimal design of clinical trials to facilitate development of ADCs for the treatment of gliomas and brain metastases.
Collapse
Affiliation(s)
- Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria
| | - Rupert Bartsch
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Emilie Le Rhun
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria
| | - Priscilla K Brastianos
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quirónsalud Group, Madrid and Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
- Medical Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, VIC, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew B Lassman
- Division of Neuro-Oncology, Department of Neurology, Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Hospital, New York, NY, USA
| | - Patrick Y Wen
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Martin van den Bent
- The Brain Tumour Center, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Zhu Y, Liu K, Wang K, Zhu H. Treatment-related adverse events of antibody-drug conjugates in clinical trials: A systematic review and meta-analysis. Cancer 2023; 129:283-295. [PMID: 36408673 PMCID: PMC10099922 DOI: 10.1002/cncr.34507] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) have complex molecular structures and have been tested in numerous clinical trials. Therefore, understanding the mechanisms of their toxicity when applied in medical practice is of high importance. METHODS In a systematic review and meta-analysis of data gathered from different scientific databases (PubMed, Embase, Cochrane, and Web of Science) between January 1, 2000, and June 7, 2022, the authors applied a random-effects model with logit transformation and evaluated the heterogeneity between studies using I2 statistics. The primary outcome was the incidence and 95% confidence interval (CI) for all-grade and grade ≥3 treatment-related adverse events and differences between different drugs, molecular structures, and cancer types. RESULTS In total, 2511 records were identified that included 169 clinical trials involving 22,492 patients. The overall incidence of treatment-related adverse events was 91.2% (95% CI, 90.7%-91.7%; I2 = 95.9%) for all-grade adverse events and 46.1% (95% CI, 45.2%-47.0%; I2 = 96.3%) for grade ≥3 adverse events. The most common all-grade adverse events were lymphopenia (53.0%; 95% CI, 48.7%-57.3%), nausea (44.1%; 95% CI, 43.2%-44.9%), neutropenia (43.7%; 95% CI, 42.6%-44.9%), blurred vision (40.5%; 95% CI, 37.4%-43.6%), and peripheral neuropathy (39.6%; 95% CI, 38.2%-41.1%); and the most common grade ≥3 adverse events were neutropenia (31.2%; 95% CI, 30.2%-32.3%), hypoesthesia (23.3%; 95% CI, 10.6%-35.9%), thrombocytopenia (22.6%; 95% CI, 21.3%-23.9%), febrile neutropenia (21.2%; 95% CI, 19.3%-23.1%), and lymphopenia (21.0%; 95% CI, 18.2%-23.7%). CONCLUSIONS Different ADCs appear to affect various treatment-related adverse events and provide comprehensive data on treatment-related adverse events for ADCs. The current results provide an important reference for clinicians and patients on how to care for toxicities from ADCs in clinical practice. LAY SUMMARY Unique anticancer drugs called antibody-drug conjugates (ADCs) have made significant progress in oncology in recent years because of their great success, and they are rapidly being used in the clinic as well as in hundreds of ongoing trials exploring their further use. The occurrence of serious side effects (adverse events) related to the receipt of ADCs was studied using data from 169 clinical trials involving 22,492 patients to determine the treatment-related causes of higher toxicity and adverse events in patients who receive ADCs, because these data are crucial for informing physicians how to safely treat patients using ADCs. The results indicate that different ADCs appear to affect various adverse events related to their use, providing comprehensive data on these ADCs that provide an important reference for clinicians and patients on how to care for toxicities from ADCs in clinical practice.
Collapse
Affiliation(s)
- Youwen Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kun Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kailing Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Abstract
Glioblastoma is the most aggressive primary brain tumor with a poor prognosis. The 2021 WHO CNS5 classification has further stressed the importance of molecular signatures in diagnosis although therapeutic breakthroughs are still lacking. In this review article, updates on the current and novel therapies in IDH-wildtype GBM will be discussed.
Collapse
Affiliation(s)
- Jawad M Melhem
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - James R Perry
- Division of Neurology, Department of Medicine, Faculty of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
9
|
Kong BY, Sim HW, Barnes EH, Nowak AK, Hovey EJ, Jeffree R, Harrup R, Parkinson J, Gan HK, Pinkham MB, Yip S, Hall M, Tu E, Carter C, Koh ES, Lwin Z, Dowling A, Simes JS, Gedye C. Multi-Arm GlioblastoMa Australasia (MAGMA): protocol for a multiarm randomised clinical trial for people affected by glioblastoma. BMJ Open 2022; 12:e058107. [PMID: 36104135 PMCID: PMC10441685 DOI: 10.1136/bmjopen-2021-058107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 08/12/2022] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common malignant primary central nervous system cancer in adults. The objective of the Multi-Arm GlioblastoMa Australasia (MAGMA) trial is to test hypotheses in real world setting to improve survival of people with GBM. Initial experimental arms are evaluating the effectiveness of interventions in newly diagnosed GBM (ndGBM). This study will compare maximal surgical resection followed by chemoradiotherapy plus adjuvant chemotherapy for 6 months with the addition of (1) 'neoadjuvant' chemotherapy beginning as soon as possible after surgery and/or (2) adjuvant chemotherapy continued until progression within the same study platform. METHODS AND ANALYSIS MAGMA will establish a platform for open-label, multiarm, multicentre randomised controlled testing of treatments for GBM. The study began recruiting in September 2020 and recruitment to the initial two interventions in MAGMA is expected to continue until September 2023.Adults aged ≥18 years with ndGBM will be given the option of undergoing randomisation to each study intervention separately, thereby giving rise to a partial factorial design, with two separate randomisation time points, one for neoadjuvant therapy and one for extended therapy. Patients will have the option of being randomised at each time point or continuing on with standard treatment.The primary outcome for the study is overall survival from the date of initial surgery until death from any cause. Secondary outcomes include progression-free survival, time to first non-temozolomide treatment, overall survival from each treatment randomisation, clinically significant toxicity as measured by grade 3 or 4 adverse events and health-related quality-of-life measures. Tertiary outcomes are predictive/prognostic biomarkers and health utilities and incremental cost-effectiveness ratio.The primary analysis of overall survival will be performed separately for each study intervention according to the intention to treat principle on all patients randomised to each study intervention. ETHICS AND DISSEMINATION The study (Protocol version 2.0 dated 23 November 2020) was approved by a lead Human Research Ethics Committee (Sydney Local Health District: 2019/ETH13297). The study will be conducted in accordance with the principles of the Declaration of Helsinki and Good Clinical Practice. TRIAL REGISTRATION NUMBER ACTRN12620000048987.
Collapse
Affiliation(s)
- Benjamin Y Kong
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Hao-Wen Sim
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | | | - Anna K Nowak
- Medical School, The University of Western Australia, Crawley, Western Australia, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Perth, Australia
| | - Elizabeth J Hovey
- Department of Medical Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Randwick, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Rosalind Jeffree
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Rosemary Harrup
- Cancer and Blood Services, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Jonathon Parkinson
- Department of Neurosurgery, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Hui K Gan
- Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
- Department of Medical Oncology, Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, VIC, Australia
| | - Mark B Pinkham
- Department of Radiation Oncology, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Sonia Yip
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Merryn Hall
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Emily Tu
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Candace Carter
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Eng-Siew Koh
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Radiation Oncology, Liverpool Cancer Therapy Centre, Liverpool, New South Wales, Australia
- Collaboration for Cancer Outcomes, Research and Evaluation, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Zarnie Lwin
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Anthony Dowling
- Department of Medicine, University of Melbourne Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
- Department of Medical Oncology, St Vincent's Hospital Melbourne Pty Ltd, Fitzroy, Victoria, Australia
| | - John S Simes
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Craig Gedye
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| |
Collapse
|
10
|
Functional Precision Oncology: The Next Frontier to Improve Glioblastoma Outcome? Int J Mol Sci 2022; 23:ijms23158637. [PMID: 35955765 PMCID: PMC9369403 DOI: 10.3390/ijms23158637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma remains the most malignant and intrinsically resistant brain tumour in adults. Despite intensive research over the past few decades, through which numerous potentially druggable targets have been identified, virtually all clinical trials of the past 20 years have failed to improve the outcome for the vast majority of GBM patients. The observation that small subgroups of patients displayed a therapeutic response across several unsuccessful clinical trials suggests that the GBM patient population probably consists of multiple subgroups that probably all require a distinct therapeutic approach. Due to extensive inter- and intratumoral heterogeneity, assigning the right therapy to each patient remains a major challenge. Classically, bulk genetic profiling would be used to identify suitable therapies, although the success of this approach remains limited due to tumor heterogeneity and the absence of direct relationships between mutations and therapy responses in GBM. An attractive novel strategy aims at implementing methods for functional precision oncology, which refers to the evaluation of treatment efficacies and vulnerabilities of (ex vivo) living tumor cells in a highly personalized way. Such approaches are currently being implemented for other cancer types by providing rapid, translatable information to guide patient-tailored therapeutic selections. In this review, we discuss the current state of the art of transforming technologies, tools and challenges for functional precision oncology and how these could improve therapy selection for GBM patients.
Collapse
|
11
|
Yang X, Chen Y, Li M, Zhu W. ERBB3 methylation and immune infiltration in tumor microenvironment of cervical cancer. Sci Rep 2022; 12:8112. [PMID: 35581263 PMCID: PMC9114106 DOI: 10.1038/s41598-022-11415-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/22/2022] [Indexed: 11/09/2022] Open
Abstract
ERBB3, a member of the ERBB family of receptor tyrosine kinases, plays an important role in cancer, despite its lack of intrinsic carcinogenic mechanism of cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC). Research on bioinformatics methods through multi-omics, this work proves that ERBB3 gene mutation, methylation modification have extensive regulatory mechanisms on the CESC microenvironment. We found that ERBB3 is involved in carcinogenesis of cervical cancer and is not associated with its prognosis. The carcinogenic mechanism is mainly related to the suppression of the immune system between tumor infiltrating lymphocytes (TILs) and the methylation of the RNA level. Our study indicated ERBB3 is more likely to be a carcinogenic factor than a key prognostic factor for cervical cancer. Methylation of ERBB3 may work as a checkpoint immunotherapy target in CESC, DNA methylation modification of the 4480 base pair downstream of ERBB3 transcription initiation site was the highest.
Collapse
Affiliation(s)
- Xiaoyue Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, Jiangsu, China.,Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Sanxiang Road 1055, Suzhou, 215000, Jiangsu, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Sanxiang Road 1055, Suzhou, 215000, Jiangsu, China
| | - Mei Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, Jiangsu, China.
| | - Weipei Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Sanxiang Road 1055, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
12
|
Kapoor V, Singh AK, Lewis CD, Deore S, Hallahan DE. Exploiting Radiation Induction of Antigens in Cancer: Targeted Drug Delivery. Int J Mol Sci 2022; 23:ijms23063041. [PMID: 35328459 PMCID: PMC8953554 DOI: 10.3390/ijms23063041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 11/25/2022] Open
Abstract
Therapeutic antibodies used to treat cancer are effective in patients with advanced-stage disease. For example, antibodies that activate T-lymphocytes improve survival in many cancer subtypes. In addition, antibody–drug conjugates effectively target cytotoxic agents that are specific to cancer. This review discusses radiation-inducible antigens, which are stress-regulated proteins that are over-expressed in cancer. These inducible cell surface proteins become accessible to antibody binding during the cellular response to genotoxic stress. The lead antigens are induced in all histologic subtypes and nearly all advanced-stage cancers, but show little to no expression in normal tissues. Inducible antigens are exploited by using therapeutic antibodies that bind specifically to these stress-regulated proteins. Antibodies that bind to the inducible antigens GRP78 and TIP1 enhance the efficacy of radiotherapy in preclinical cancer models. The conjugation of cytotoxic drugs to the antibodies further improves cancer response. This review focuses on the use of radiotherapy to control the cancer-specific binding of therapeutic antibodies and antibody–drug conjugates.
Collapse
Affiliation(s)
- Vaishali Kapoor
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO 63108, USA; (V.K.); (A.K.S.); (C.D.L.)
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Abhay K. Singh
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO 63108, USA; (V.K.); (A.K.S.); (C.D.L.)
| | - Calvin D. Lewis
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO 63108, USA; (V.K.); (A.K.S.); (C.D.L.)
- Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Sapna Deore
- Medical Guidance Systems LLC, St. Louis, MO 63110, USA;
| | - Dennis E. Hallahan
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO 63108, USA; (V.K.); (A.K.S.); (C.D.L.)
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63108, USA
- Correspondence: ; Tel.: +314-362-9700; Fax: +314-747-5498
| |
Collapse
|
13
|
Parakh S, Nicolazzo J, Scott AM, Gan HK. Antibody Drug Conjugates in Glioblastoma - Is There a Future for Them? Front Oncol 2021; 11:718590. [PMID: 34926242 PMCID: PMC8678283 DOI: 10.3389/fonc.2021.718590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and fatal malignancy that despite decades of trials has limited therapeutic options. Antibody drug conjugates (ADCs) are composed of a monoclonal antibody which specifically recognizes a cellular surface antigen linked to a cytotoxic payload. ADCs have demonstrated superior efficacy and/or reduced toxicity in a range of haematological and solid tumors resulting in nine ADCs receiving regulatory approval. ADCs have also been explored in patients with brain tumours but with limited success to date. While earlier generations ADCs in glioma patients have had limited success and high toxicity, newer and improved ADCs characterised by low immunogenicity and more effective payloads have shown promise in a range of tumour types. These newer ADCs have also been tested in glioma patients, however, with mixed results. Factors affecting the effectiveness of ADCs to target the CNS include the blood brain barrier which acts as a physical and biochemical barrier, the pro-cancerogenic and immunosuppressive tumor microenvironment and tumour characteristics like tumour volume and antigen expression. In this paper we review the data regarding the ongoing the development of ADCs in glioma patients as well as potential strategies to overcome these barriers to maximise their therapeutic potential.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Joseph Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Hui Kong Gan
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
14
|
Narita Y, Muragaki Y, Kagawa N, Asai K, Nagane M, Matsuda M, Ueki K, Kuroda J, Date I, Kobayashi H, Kumabe T, Beppu T, Kanamori M, Kasai S, Nishimura Y, Xiong H, Ocampo C, Yamada M, Mishima K. Safety and efficacy of depatuxizumab mafodotin in Japanese patients with malignant glioma: A nonrandomized, phase 1/2 trial. Cancer Sci 2021; 112:5020-5033. [PMID: 34609773 PMCID: PMC8645742 DOI: 10.1111/cas.15153] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
INTELLANCE‐J was a phase 1/2 study of a potent antibody‐drug conjugate targeting epidermal growth factor receptor (EGFR), depatuxizumab mafodotin (Depatux‐M), as a second‐ or first‐line therapy, alone or combined with chemotherapy or chemoradiotherapy in 53 Japanese patients with World Health Organization (WHO) grade III/IV glioma. In second‐line arms, patients with EGFR‐amplified recurrent WHO grade III/IV glioma received Depatux‐M plus chemotherapy (temozolomide) or Depatux‐M alone regardless of EGFR status. In first‐line arms, patients with newly diagnosed WHO grade III/IV glioma received Depatux‐M plus chemoradiotherapy. The study was halted following lack of survival benefit with first‐line Depatux‐M in the global trial INTELLANCE‐1. The primary endpoint was 6‐month progression‐free survival (PFS) in patients with EGFR‐amplified tumors receiving second‐line Depatux‐M plus chemotherapy. Common nonocular treatment‐emergent adverse events (TEAEs) with both second‐line and first‐line Depatux‐M included lymphopenia (42%, 33%, respectively), thrombocytopenia (39%, 47%), alanine aminotransferase increase (29%, 47%), and aspartate aminotransferase increase (24%, 60%); incidence of grade ≥3 TEAEs was 66% and 53%, respectively. Ocular side effects (OSEs) occurred in 93% of patients receiving second‐line Depatux‐M plus chemotherapy and all patients receiving second‐line Depatux‐M alone or first‐line Depatux‐M plus chemoradiotherapy. Most OSEs were manageable with dose modifications and concomitant medications. The 6‐month PFS estimate was 25.6% (95% confidence interval [CI] 11.4‒42.6), and median PFS was 2.1 months (95% CI 1.9‒3.9) with second‐line Depatux‐M plus chemotherapy in the EGFR‐amplified subgroup. This study showed acceptable safety profile of Depatux‐M alone or plus chemotherapy/chemoradiotherapy in Japanese patients with WHO grade III/IV glioma. The study was registered at ClinicalTrials.gov (NCT02590263).
Collapse
Affiliation(s)
- Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshihiro Muragaki
- Department of Neurosurgery, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Naoki Kagawa
- Department of Neurosurgery, Osaka University Hospital, Osaka, Japan
| | - Katsunori Asai
- Department of Neurosurgery, Osaka International Cancer Institute, Osaka, Japan
| | - Motoo Nagane
- Faculty of Medicine, Department of Neurosurgery, Kyorin University, Tokyo, Japan
| | - Masahide Matsuda
- Department of Neurosurgery, University of Tsukuba, Ibaraki, Japan
| | - Keisuke Ueki
- Department of Neurosurgery, Dokkyo Medical University Hospital, Tochigi, Japan
| | - Junichiro Kuroda
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Isao Date
- Department of Neurosurgery, Okayama University Hospital, Okayama, Japan
| | - Hiroyuki Kobayashi
- Department of Neurosurgery, Hokkaido University Hospital, Hokkaido, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University Hospital, Kanagawa, Japan
| | - Takaaki Beppu
- Department of Neurosurgery, Iwate Medical University Hospital, Iwate, Japan
| | - Masayuki Kanamori
- Department of Neurosurgery, Tohoku University Hospital, Miyagi, Japan
| | | | | | - Hao Xiong
- AbbVie Inc., North Chicago, Illinois, USA
| | | | - Masakazu Yamada
- Department of Ophthalmology, Kyorin University Hospital, Tokyo, Japan
| | - Kazuhiko Mishima
- Department of Neuro-Oncology/Neurosurgery, International Medical Center, Saitama Medical University, Saitama, Japan
| |
Collapse
|
15
|
Thokala R, Binder ZA, Yin Y, Zhang L, Zhang JV, Zhang DY, Milone MC, Ming GL, Song H, O'Rourke DM. High-Affinity Chimeric Antigen Receptor With Cross-Reactive scFv to Clinically Relevant EGFR Oncogenic Isoforms. Front Oncol 2021; 11:664236. [PMID: 34568006 PMCID: PMC8461175 DOI: 10.3389/fonc.2021.664236] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/18/2021] [Indexed: 12/31/2022] Open
Abstract
Tumor heterogeneity is a key reason for therapeutic failure and tumor recurrence in glioblastoma (GBM). Our chimeric antigen receptor (CAR) T cell (2173 CAR T cells) clinical trial (NCT02209376) against epidermal growth factor receptor (EGFR) variant III (EGFRvIII) demonstrated successful trafficking of T cells across the blood–brain barrier into GBM active tumor sites. However, CAR T cell infiltration was associated only with a selective loss of EGFRvIII+ tumor, demonstrating little to no effect on EGFRvIII- tumor cells. Post-CAR T-treated tumor specimens showed continued presence of EGFR amplification and oncogenic EGFR extracellular domain (ECD) missense mutations, despite loss of EGFRvIII. To address tumor escape, we generated an EGFR-specific CAR by fusing monoclonal antibody (mAb) 806 to a 4-1BB co-stimulatory domain. The resulting construct was compared to 2173 CAR T cells in GBM, using in vitro and in vivo models. 806 CAR T cells specifically lysed tumor cells and secreted cytokines in response to amplified EGFR, EGFRvIII, and EGFR-ECD mutations in U87MG cells, GBM neurosphere-derived cell lines, and patient-derived GBM organoids. 806 CAR T cells did not lyse fetal brain astrocytes or primary keratinocytes to a significant degree. They also exhibited superior antitumor activity in vivo when compared to 2173 CAR T cells. The broad specificity of 806 CAR T cells to EGFR alterations gives us the potential to target multiple clones within a tumor and reduce opportunities for tumor escape via antigen loss.
Collapse
Affiliation(s)
- Radhika Thokala
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yibo Yin
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Logan Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jiasi Vicky Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y Zhang
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Biochemistry and Molecular Physics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael C Milone
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-Li Ming
- Biochemistry and Molecular Physics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
16
|
Gan HK, Parakh S, Lassman AB, Seow A, Lau E, Lee ST, Ameratunga M, Perchyonok Y, Cao D, Burvenich IJG, O'Keefe GJ, Rigopoulos A, Gomez E, Maag D, Scott AM. Tumor volumes as a predictor of response to the anti-EGFR antibody drug conjugate depatuxizumab mafadotin. Neurooncol Adv 2021; 3:vdab102. [PMID: 34549181 PMCID: PMC8446913 DOI: 10.1093/noajnl/vdab102] [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] [Indexed: 11/13/2022] Open
Abstract
Background The adverse impact of increasing brain tumor size on the efficacy of antibody-drug conjugates (ADCs) was investigated preclinically then validated with clinical data. Methods—Preclinical study The impact of tumor size on ADC tumor delivery and treatment response was evaluated in an EGFR-amplified patient-derived glioblastoma (GBM) model following treatment with Depatuxizumab mafadotin (Depatux-M). Biodistribution and imaging studies correlated drug distribution with starting treatment volume and anti-tumor activity. Methods—Clinical study M12-356 was a Phase I study of Depatux-M in patients with GBM. Blinded volumetric analysis of baseline tumor volumes of M12-356 patients was undertaken by two reviewers and results correlated with response and survival. Results Preclinically, imaging and biodistribution studies showed specific and significantly higher tumor uptake of zirconium-89 labeled Depatux-M (89Zr-Depatux-M) in mice with smaller tumor volume (~98 mm3) versus those with larger volumes (~365 mm3); concordantly, mice with tumor volumes ≤100 mm3 at treatment commencement had significantly better growth inhibition by Depatux-M (93% vs 27%, P < .001) and significantly longer overall survival (P < .0001) compared to tumors ≥400 mm3. Clinically, patients with tumor volumes <25 cm3 had significantly higher response rates (17% vs. 0%, P = .009) and longer overall survival (0.5 vs 0.89 years, P = .001) than tumors above 25 cm3. Conclusion Both preclinical and clinical data showed intra-tumoral concentration and efficacy of Depatux-m inversely correlated with tumor size. This finding merit further investigation with pretreatment tumor volume as a predictor for response to ADCs, in both gliomas and other solid tumors.
Collapse
Affiliation(s)
- Hui K Gan
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia.,Department of Medicine, University of Melbourne, Parkville, Australia
| | - Sagun Parakh
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medical Oncology, Monash Health, Clayton, Melbourne, Australia
| | - Andrew B Lassman
- Division of Neuro-Oncology, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Aidan Seow
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| | - Eddie Lau
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia.,Department of Radiology, Austin Health, Heidelberg, Melbourne, Australia
| | - Sze Ting Lee
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medicine, University of Melbourne, Parkville, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| | - Malaka Ameratunga
- Department of Medical Oncology, Austin Health, Heidelberg, Melbourne, Australia
| | - Yuliya Perchyonok
- Department of Radiology, Austin Health, Heidelberg, Melbourne, Australia
| | - Diana Cao
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia
| | - Ingrid J G Burvenich
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia
| | - Graeme J O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| | - Angela Rigopoulos
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia
| | - Erica Gomez
- Research and Development Department, AbbVie Inc., North Chicago, Illinois, USA
| | - David Maag
- Research and Development Department, AbbVie Inc., North Chicago, Illinois, USA
| | - Andrew M Scott
- Tumour Targeting Program, Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Melbourne, Australia.,La Trobe University School of Cancer Medicine, Heidelberg, Melbourne, Australia.,Department of Medicine, University of Melbourne, Parkville, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Melbourne, Australia
| |
Collapse
|
17
|
PD-L1 Expression Fluctuates Concurrently with Cyclin D in Glioblastoma Cells. Cells 2021; 10:cells10092366. [PMID: 34572014 PMCID: PMC8468141 DOI: 10.3390/cells10092366] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Despite Glioblastoma (GBM) frequently expressing programmed cell death ligand-1 (PD-L1), treatment with anti-programmed cell death-1 (PD1) has not yielded brilliant results. Intratumor variability of PD-L1 can impact determination accuracy. A previous study on mouse embryonic fibroblasts (MEFs) reported a role for cyclin-D in control of PD-L1 expression. Because tumor-cell growth within a cancer is highly heterogeneous, we looked at whether PD-L1 and its cochaperone FKBP51s were influenced by cell proliferation, using U251 and SF767 GBM-cell-lines. PD-L1 was measured by Western blot, flow cytometry, confocal-microscopy, quantitative PCR (qPCR), CCND1 by qPCR, FKBP51s by Western blot and confocal-microscopy. Chromatin-Immunoprecipitation assay (xChIp) served to assess the DNA-binding of FKBP51 isoforms. In the course of cell culture, PD-L1 appeared to increase concomitantly to cyclin-D on G1/S transition, to decrease during exponential cell growth progressively. We calculated a correlation between CCND1 and PD-L1 gene expression levels. In the temporal window of PD-L1 and CCND1 peak, FKBP51s localized in ER. When cyclin-D declined, FKBP51s went nuclear. XChIp showed that FKBP51s binds CCND1 gene in a closed-chromatin configuration. Our finding suggests that the dynamism of PD-L1 expression in GBM follows cyclin-D fluctuation and raises the hypothesis that FKBP51s might participate in the events that govern cyclin-D oscillation.
Collapse
|
18
|
Bolcaen J, Nair S, Driver CHS, Boshomane TMG, Ebenhan T, Vandevoorde C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals (Basel) 2021; 14:626. [PMID: 34209513 PMCID: PMC8308832 DOI: 10.3390/ph14070626] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) remains the most fatal brain tumor characterized by a high infiltration rate and treatment resistance. Overexpression and/or mutation of receptor tyrosine kinases is common in GB, which subsequently leads to the activation of many downstream pathways that have a critical impact on tumor progression and therapy resistance. Therefore, receptor tyrosine kinase inhibitors (RTKIs) have been investigated to improve the dismal prognosis of GB in an effort to evolve into a personalized targeted therapy strategy with a better treatment outcome. Numerous RTKIs have been approved in the clinic and several radiopharmaceuticals are part of (pre)clinical trials as a non-invasive method to identify patients who could benefit from RTKI. The latter opens up the scope for theranostic applications. In this review, the present status of RTKIs for the treatment, nuclear imaging and targeted radionuclide therapy of GB is presented. The focus will be on seven tyrosine kinase receptors, based on their central role in GB: EGFR, VEGFR, MET, PDGFR, FGFR, Eph receptor and IGF1R. Finally, by way of analyzing structural and physiological characteristics of the TKIs with promising clinical trial results, four small molecule RTKIs were selected based on their potential to become new therapeutic GB radiopharmaceuticals.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Cathryn H. S. Driver
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Brits 0240, South Africa;
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Preclinical Drug Development Platform, Department of Science and Technology, North West University, Potchefstroom 2520, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| |
Collapse
|
19
|
Van Den Bent M, Eoli M, Sepulveda JM, Smits M, Walenkamp A, Frenel JS, Franceschi E, Clement PM, Chinot O, De Vos F, Whenham N, Sanghera P, Weller M, Dubbink HJ, French P, Looman J, Dey J, Krause S, Ansell P, Nuyens S, Spruyt M, Brilhante J, Coens C, Gorlia T, Golfinopoulos V. INTELLANCE 2/EORTC 1410 randomized phase II study of Depatux-M alone and with temozolomide vs temozolomide or lomustine in recurrent EGFR amplified glioblastoma. Neuro Oncol 2021; 22:684-693. [PMID: 31747009 PMCID: PMC7229258 DOI: 10.1093/neuonc/noz222] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Depatuxizumab mafodotin (Depatux-M) is a tumor-specific antibody–drug conjugate consisting of an antibody (ABT-806) directed against activated epidermal growth factor receptor (EGFR) and the toxin monomethylauristatin-F. We investigated Depatux-M in combination with temozolomide or as a single agent in a randomized controlled phase II trial in recurrent EGFR amplified glioblastoma. Methods Eligible were patients with centrally confirmed EGFR amplified glioblastoma at first recurrence after chemo-irradiation with temozolomide. Patients were randomized to either Depatux-M 1.25 mg/kg every 2 weeks intravenously, or this treatment combined with temozolomide 150–200 mg/m2 day 1–5 every 4 weeks, or either lomustine or temozolomide. The primary endpoint of the study was overall survival. Results Two hundred sixty patients were randomized. In the primary efficacy analysis with 199 events (median follow-up 15.0 mo), the hazard ratio (HR) for the combination arm compared with the control arm was 0.71 (95% CI = 0.50, 1.02; P = 0.062). The efficacy of Depatux-M monotherapy was comparable to that of the control arm (HR = 1.04, 95% CI = 0.73, 1.48; P = 0.83). The most frequent toxicity in Depatux-M treated patients was a reversible corneal epitheliopathy, occurring as grades 3–4 adverse events in 25–30% of patients. In the long-term follow-up analysis with median follow-up of 28.7 months, the HR for the comparison of the combination arm versus the control arm was 0.66 (95% CI = 0.48, 0.93). Conclusion This trial suggests a possible role for the use of Depatux-M in combination with temozolomide in EGFR amplified recurrent glioblastoma, especially in patients relapsing well after the end of first-line adjuvant temozolomide treatment. (NCT02343406)
Collapse
Affiliation(s)
- Martin Van Den Bent
- Brain Tumor Institute Erasmus Medical Center (MC) Cancer Institute, Rotterdam, the Netherlands
| | - Marica Eoli
- Department of Neurology, Carlo Besta Institute, Milan, Italy
| | | | - Marion Smits
- Department of Radiology, Erasmus MC, Rotterdam, the Netherlands
| | - Annemiek Walenkamp
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jean-Sebastian Frenel
- Department of Medical Oncology, René Gauducheau Center for Cancer Care, Nantes, France
| | - Enrico Franceschi
- Department of Medical Oncology, Local Health Unit Agency/Scientific Institute for Research, Hospitalization, and Healthcare (AUSL/IRCCS) Neurological Sciences, Bologna, Italy
| | - Paul M Clement
- Department of Medical Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Olivier Chinot
- Department of Neuro-Oncology, Institute of Neurophysiopathology, Aix-Marseille University, Marseille, France
| | - Filip De Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Nicolas Whenham
- Department of Medical Oncology, European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | | | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - H J Dubbink
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Pim French
- Brain Tumor Institute Erasmus Medical Center (MC) Cancer Institute, Rotterdam, the Netherlands
| | | | - Jyotirmoy Dey
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.,Abbvie, Chicago, IL USA
| | | | | | | | - Maarten Spruyt
- Brain Tumor Institute Erasmus Medical Center (MC) Cancer Institute, Rotterdam, the Netherlands
| | | | | | | | | |
Collapse
|
20
|
Chen L, Qin D, Guo X, Wang Q, Li J. Putting Proteomics Into Immunotherapy for Glioblastoma. Front Immunol 2021; 12:593255. [PMID: 33708196 PMCID: PMC7940695 DOI: 10.3389/fimmu.2021.593255] [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: 08/10/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
In glioblastoma, the most aggressive brain cancer, a complex microenvironment of heterogeneity and immunosuppression, are considerable hurdles to classify the subtypes and promote treatment progression. Treatments for glioblastoma are similar to standard therapies for many other cancers and do not effectively prolong the survival of patients, due to the unique location and heterogeneous characteristics of glioblastoma. Immunotherapy has shown a promising effect for many other tumors, but its application for glioma still has some challenges. The recent breakthrough of high-throughput liquid chromatography-mass spectrometry (LC-MS/MS) systems has allowed researchers to update their strategy for identifying and quantifying thousands of proteins in a much shorter time with lesser effort. The protein maps can contribute to generating a complete map of regulatory systems to elucidate tumor mechanisms. In particular, newly developed unicellular proteomics could be used to determine the microenvironment and heterogeneity. In addition, a large scale of differentiated proteins provides more ways to precisely classify tumor subtypes and construct a larger library for biomarkers and biotargets, especially for immunotherapy. A series of advanced proteomic studies have been devoted to the different aspects of immunotherapy for glioma, including monoclonal antibodies, oncolytic viruses, dendritic cell (DC) vaccines, and chimeric antigen receptor (CAR) T cells. Thus, the application of proteomics in immunotherapy may accelerate research on the treatment of glioblastoma. In this review, we evaluate the frontline applications of proteomics strategies for immunotherapy in glioblastoma research.
Collapse
Affiliation(s)
- Liangyu Chen
- Department of Proteomics, Tianjin Enterprise Key Laboratory of Clinical Multi-omics, Tianjin, China
| | - Di Qin
- Department of Proteomics, Tianjin Enterprise Key Laboratory of Clinical Multi-omics, Tianjin, China
| | - Xinyu Guo
- Department of Proteomics, Tianjin Enterprise Key Laboratory of Clinical Multi-omics, Tianjin, China
| | - Qixue Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
| | - Jie Li
- Department of Proteomics, Tianjin Enterprise Key Laboratory of Clinical Multi-omics, Tianjin, China
| |
Collapse
|
21
|
Liu JY, Fu WQ, Zheng XJ, Li W, Ren LW, Wang JH, Yang C, Du GH. Avasimibe exerts anticancer effects on human glioblastoma cells via inducing cell apoptosis and cell cycle arrest. Acta Pharmacol Sin 2021; 42:97-107. [PMID: 32451414 PMCID: PMC7921416 DOI: 10.1038/s41401-020-0404-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/19/2020] [Indexed: 01/11/2023]
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults, but there is no effective drug available for GBM. Avasimibe is a potent inhibitor of acyl-coenzyme A: cholesterol acyltransferase-1 (ACAT-1), which was used to treat atherosclerosis. Experimental evidence and bioinformatics have shown that avasimibe has anticancer activity. In this study we investigated the anticancer effects of avasimibe on human glioblastoma cells and the underlying mechanisms. Our results showed that avasimibe dose-dependently inhibited the proliferation of U251 and U87 human glioblastoma cells with IC50 values of 20.29 and 28.27 μM, respectively, at 48 h. Avasimibe (7.5, 15, 30 μM) decreased the DNA synthesis, and inhibited the colony formation of the tumor cells. Treatment of avasimibe also dose-dependently increased the apoptotic rate of tumor cells, decreased the mitochondrial membrane potential, induced the activity of caspase-3/7, and increased the protein expression of cleaved caspase-9, cleaved PARP and Bax in U251 and U87 cells. RNA-sequencing analyses revealed that avasimibe suppressed the expression of CDK2, cyclin E1, CDK4, cyclin D, CDK1, cyclin B1, Aurora A, and PLK1, while induced the expression of p53, p21, p27, and GADD45A, which was validated by Western blot analysis. These results demonstrated that avasimibe induced mitochondria-dependent apoptosis in glioblastoma cells, which was associated with arresting the cell cycle at G0/G1 phase and G2/M phase by regulating the p53/p21 pathway, p53/GADD45A and Aurora A/PLK1 signaling pathways. In U87 xenograft nude mice model, administration of avasimibe (15, 30 mg·kg-1·d-1, ip, for 18 days) dose-dependently inhibit the tumor growth. Taken together, our results demonstrated that avasimibe might be a promising chemotherapy drug in the treatment of GBM.
Collapse
Affiliation(s)
- Jin-Yi Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wei-Qi Fu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Xiang-Jin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Li-Wen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Jin-Hua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China.
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| | - Cui Yang
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650500, China.
| | - Guan-Hua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China.
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
22
|
Farrell C, Shi W, Bodman A, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of emerging developments in the management of newly diagnosed glioblastoma. J Neurooncol 2020; 150:269-359. [PMID: 33215345 DOI: 10.1007/s11060-020-03607-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022]
Abstract
TARGET POPULATION These recommendations apply to adult patients with newly diagnosed or suspected glioblastoma. IMAGING Question What imaging modalities are in development that may be able to provide improvements in diagnosis, and therapeutic guidance for individuals with newly diagnosed glioblastoma? RECOMMENDATION Level III: It is suggested that techniques utilizing magnetic resonance imaging for diffusion weighted imaging, and to measure cerebral blood and magnetic spectroscopic resonance imaging of N-acetyl aspartate, choline and the choline to N-acetyl aspartate index to assist in diagnosis and treatment planning in patients with newly diagnosed or suspected glioblastoma. SURGERY Question What new surgical techniques can be used to provide improved tumor definition and resectability to yield better tumor control and prognosis for individuals with newly diagnosed glioblastoma? RECOMMENDATIONS Level II: The use of 5-aminolevulinic acid is recommended to improve extent of tumor resection in patients with newly diagnosed glioblastoma. Level II: The use of 5-aminolevulinic acid is recommended to improve median survival and 2 year survival in newly diagnosed glioblastoma patients with clinical characteristics suggesting poor prognosis. Level III: It is suggested that, when available, patients be enrolled in properly designed clinical trials assessing the value of diffusion tensor imaging in improving the safety of patients with newly diagnosed glioblastoma undergoing surgery. NEUROPATHOLOGY Question What new pathology techniques and measurement of biomarkers in tumor tissue can be used to provide improved diagnostic ability, and determination of therapeutic responsiveness and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATIONS Level II: Assessment of tumor MGMT promoter methylation status is recommended as a significant predictor of a longer progression free survival and overall survival in patients with newly diagnosed with glioblastoma. Level II: Measurement of tumor expression of neuron-glia-2, neurofilament protein, glutamine synthetase and phosphorylated STAT3 is recommended as a predictor of overall survival in patients with newly diagnosed with glioblastoma. Level III: Assessment of tumor IDH1 mutation status is suggested as a predictor of longer progression free survival and overall survival in patients with newly diagnosed with glioblastoma. Level III: Evaluation of tumor expression of Phosphorylated Mitogen-Activated Protein Kinase protein, EGFR protein, and Insulin-like Growth Factor-Binding Protein-3 is suggested as a predictor of overall survival in patients with newly diagnosed with glioblastoma. RADIATION Question What radiation therapy techniques are in development that may be used to provide improved tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATIONS Level III: It is suggested that patients with newly diagnosed glioblastoma undergo pretreatment radio-labeled amino acid tracer positron emission tomography to assess areas at risk for tumor recurrence to assist in radiation treatment planning. Level III: It is suggested that, when available, patients be with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of radiation dose escalation, altered fractionation, or new radiation delivery techniques. CHEMOTHERAPY Question What emerging chemotherapeutic agents or techniques are available to provide better tumor control and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no emerging chemotherapeutic agents or techniques were identified in this review that improved tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of chemotherapy. MOLECULAR AND TARGETED THERAPY Question What new targeted therapy agents are available to provide better tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no new molecular and targeted therapies have clearly provided better tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of molecular and targeted therapies IMMUNOTHERAPY: Question What emerging immunotherapeutic agents or techniques are available to provide better tumor control and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no immunotherapeutic agents have clearly provided better tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of immunologically-based therapies. NOVEL THERAPIES Question What novel therapies or techniques are in development to provide better tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATIONS Level II: The use of tumor-treating fields is recommended for patients with newly diagnosed glioblastoma who have undergone surgical debulking and completed concurrent chemoradiation without progression of disease at the time of tumor-treating field therapy initiation. Level II: It is suggested that, when available, enrollment in properly designed studies of vector containing herpes simplex thymidine kinase gene and prodrug therapies be considered in patients with newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Christopher Farrell
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wenyin Shi
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
23
|
Parrozzani R, Lombardi G, Midena E, Londei D, Padovan M, Marchione G, Caccese M, Midena G, Zagonel V, Frizziero L. Ocular Side Effects of EGFR-Inhibitor ABT-414 in Recurrent Glioblastoma: A Long-Term Safety Study. Front Oncol 2020; 10:593461. [PMID: 33154952 PMCID: PMC7591744 DOI: 10.3389/fonc.2020.593461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/28/2020] [Indexed: 11/27/2022] Open
Abstract
This study aimed to prospectively evaluate, on a long-term basis, corneal side effects secondary to compassionate administration of epidermal growth factor receptor (EGFR) inhibitor depatuxizumab mafodotin (ABT-414) in patients affected by EGFR-amplified recurrent glioblastoma. Fifteen patients with a median follow-up of 4.3 months after treatment discontinuation were enrolled. Each patient underwent full ophthalmologic examination including in vivo corneal confocal microscopy (CCM). No CTCAE grade 4 toxicity and four (27%) grade 3 toxicities were documented during treatment. Ocular symptoms (blurred vision, eye pain, photophobia) were experienced by all patients, reaching maximal severity after the second ABT-414 infusion, with persistence until treatment discontinuation. During treatment, CCM documented specific changes in the corneal epithelium and in the sub-basal nerve plexus layer fibers of all eyes. The median time of symptoms resolution after treatment discontinuation ranged from 38 days (eye pain) to 53 days (photophobia). The median time of signs resolution ranges from 14 days (corneal ulcer) to 38 days (superficial punctate epitheliopathy, corneal stroma edema and intraepithelial cysts). ABT-414 corneal side effects are detectable in all treated patients. Related symptoms are gradually experienced by all patients during treatment and although reversible, they are characterized by a relative prolonged persistence after treatment discontinuation.
Collapse
Affiliation(s)
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology-IRCCS, Padua, Italy
| | - Edoardo Midena
- Department of Ophthalmology, University of Padova, Padova, Italy.,IRCCS-Fondazione Bietti, Rome, Italy
| | - Davide Londei
- Department of Ophthalmology, University of Padova, Padova, Italy
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology-IRCCS, Padua, Italy
| | - Giulia Marchione
- Department of Ophthalmology, University of Padova, Padova, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology-IRCCS, Padua, Italy
| | - Giulia Midena
- Unità Operativa Complessa Oftalmologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology-IRCCS, Padua, Italy
| | | |
Collapse
|
24
|
Massey SC, Urcuyo JC, Marin BM, Sarkaria JN, Swanson KR. Quantifying Glioblastoma Drug Response Dynamics Incorporating Treatment Sensitivity and Blood Brain Barrier Penetrance From Experimental Data. Front Physiol 2020; 11:830. [PMID: 32973540 PMCID: PMC7472531 DOI: 10.3389/fphys.2020.00830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Many drugs investigated for the treatment of glioblastoma (GBM) have had disappointing clinical trial results. Efficacy of these agents is dependent on adequate delivery to sensitive tumor cell populations, which is limited by the blood-brain barrier (BBB). Additionally, tumor heterogeneity can lead to subpopulations of cells with different sensitivities to anti-cancer drugs, further impacting therapeutic efficacy. Thus, it may be important to evaluate the extent to which BBB limitations and heterogeneous sensitivity each contribute to a drug's failure. To address this challenge, we developed a minimal mathematical model to characterize these elements of overall drug response, informed by time-series bioluminescence imaging data from a treated patient-derived xenograft (PDX) experimental model. By fitting this mathematical model to a preliminary dataset in a series of nonlinear regression steps, we estimated parameter values for individual PDX subjects that correspond to the dynamics seen in experimental data. Using these estimates as a guide for parameter ranges, we ran model simulations and performed a parameter sensitivity analysis using Latin hypercube sampling and partial rank correlation coefficients. Results from this analysis combined with simulations suggest that BBB permeability may play a slightly greater role in therapeutic efficacy than relative drug sensitivity. Additionally, we discuss recommendations for future experiments based on insights gained from this model. Further research in this area will be vital for improving the development of effective new therapies for glioblastoma patients.
Collapse
Affiliation(s)
| | - Javier C. Urcuyo
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Bianca Maria Marin
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Kristin R. Swanson
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
- Department of Neurological Surgery, Mayo Clinic, Phoenix, AZ, United States
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
25
|
Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, Cloughesy TF, DeGroot JF, Galanis E, Gilbert MR, Hegi ME, Horbinski C, Huang RY, Lassman AB, Le Rhun E, Lim M, Mehta MP, Mellinghoff IK, Minniti G, Nathanson D, Platten M, Preusser M, Roth P, Sanson M, Schiff D, Short SC, Taphoorn MJB, Tonn JC, Tsang J, Verhaak RGW, von Deimling A, Wick W, Zadeh G, Reardon DA, Aldape KD, van den Bent MJ. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol 2020; 22:1073-1113. [PMID: 32328653 PMCID: PMC7594557 DOI: 10.1093/neuonc/noaa106] [Citation(s) in RCA: 703] [Impact Index Per Article: 140.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastomas are the most common form of malignant primary brain tumor and an important cause of morbidity and mortality. In recent years there have been important advances in understanding the molecular pathogenesis and biology of these tumors, but this has not translated into significantly improved outcomes for patients. In this consensus review from the Society for Neuro-Oncology (SNO) and the European Association of Neuro-Oncology (EANO), the current management of isocitrate dehydrogenase wildtype (IDHwt) glioblastomas will be discussed. In addition, novel therapies such as targeted molecular therapies, agents targeting DNA damage response and metabolism, immunotherapies, and viral therapies will be reviewed, as well as the current challenges and future directions for research.
Collapse
Affiliation(s)
- Patrick Y Wen
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Eudocia Quant Lee
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jill S Barnholtz-Sloan
- Case Western Reserve University School of Medicine and University Hospitals of Cleveland, Cleveland, Ohio, USA
| | - Floris P Barthel
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Tracy T Batchelor
- Department of Neurology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Susan M Chang
- University of California San Francisco, San Francisco, California, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy F Cloughesy
- David Geffen School of Medicine, Department of Neurology, University of California Los Angeles, Los Angeles, California, USA
| | - John F DeGroot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Monika E Hegi
- Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raymond Y Huang
- Division of Neuroradiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, NewYork-Presbyterian Hospital/Columbia University Irving Medical Center, New York, New York, USA
| | - Emilie Le Rhun
- University of Lille, Inserm, Neuro-oncology, General and Stereotaxic Neurosurgery service, University Hospital of Lille, Lille, France; Breast Cancer Department, Oscar Lambret Center, Lille, France and Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - David Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Heidelberg, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Patrick Roth
- Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marc Sanson
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière – Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - David Schiff
- University of Virginia School of Medicine, Division of Neuro-Oncology, Department of Neurology, University of Virginia, Charlottesville, Virginia, USA
| | - Susan C Short
- Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, UK
| | - Martin J B Taphoorn
- Department of Neurology, Medical Center Haaglanden, The Hague and Department of Neurology, Leiden University Medical Center, the Netherlands
| | | | - Jonathan Tsang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Roel G W Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Andreas von Deimling
- Neuropathology and Clinical Cooperation Unit Neuropathology, University Heidelberg and German Cancer Center, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology and Neuro-oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Gelareh Zadeh
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, Canada
| | - David A Reardon
- Dana-Farber Cancer Institute, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth D Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | |
Collapse
|
26
|
Abstract
OPINION STATEMENT Malignant gliomas remain a challenging cancer to treat due to limitations in both therapeutic and efficacious options. Tumor treating fields (TTFields) have emerged as a novel, locoregional, antineoplastic treatment modality with favorable efficacy and safety being demonstrated in the most aggressive type of malignant gliomas, glioblastoma (GBM). In 2 large randomized, controlled phase 3 trials, the addition of TTFields was associated with increased overall survival when combined with adjuvant temozolomide (TMZ) chemotherapy in patients with newly diagnosed GBM (ndGBM) and comparable overall survival compared with standard chemotherapy in patients with recurrent GBM (rGBM). TTFields target cancer cells by several mechanisms of action (MoA) including suppression of proliferation, migration and invasion, disruption of DNA repair and angiogenesis, antimitotic effects, and induction of apoptosis and immunogenic cell death. Having several MoAs makes TTFields an attractive modality to combine with standard, salvage, and novel treatment regimens (e.g., radiotherapy, chemotherapy, and immunotherapy). Treatment within the field of malignant gliomas is evolving to emphasize combinatorial approaches that work synergistically to improve patient outcomes. Here, we review the current use of TTFields in GBM, discuss MOA and treatment delivery, and consider the potential for its wider adoption in other gliomas.
Collapse
|
27
|
Farooq AV, Degli Esposti S, Popat R, Thulasi P, Lonial S, Nooka AK, Jakubowiak A, Sborov D, Zaugg BE, Badros AZ, Jeng BH, Callander NS, Opalinska J, Baron J, Piontek T, Byrne J, Gupta I, Colby K. Corneal Epithelial Findings in Patients with Multiple Myeloma Treated with Antibody-Drug Conjugate Belantamab Mafodotin in the Pivotal, Randomized, DREAMM-2 Study. Ophthalmol Ther 2020; 9:889-911. [PMID: 32712806 PMCID: PMC7708586 DOI: 10.1007/s40123-020-00280-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Patients with relapsed or refractory multiple myeloma (RRMM) represent an unmet clinical need. Belantamab mafodotin (belamaf; GSK2857916) is a first-in-class antibody-drug conjugate (ADC; or immunoconjugate) that delivers a cytotoxic payload, monomethyl auristatin F (MMAF), to myeloma cells. In the phase II DREAMM-2 study (NCT03525678), single-agent belamaf (2.5 mg/kg) demonstrated clinically meaningful anti-myeloma activity (overall response rate 32%) in patients with heavily pretreated disease. Microcyst-like epithelial changes (MECs) were common, consistent with reports from other MMAF-containing ADCs. METHODS Corneal examination findings from patients in DREAMM-2 were reviewed, and the clinical descriptions and accompanying images (slit lamp microscopy and in vivo confocal microscopy [IVCM]) of representative events were selected. A literature review on corneal events reported with other ADCs was performed. RESULTS In most patients receiving single-agent belamaf (72%; 68/95), MECs were observed by slit lamp microscopy early in treatment (69% had their first event by dose 4). However, IVCM revealed hyperreflective material. Blurred vision (25%) and dry eye (15%) were commonly reported symptoms. Management of MECs included dose delays (47%)/reductions (25%), with few patients discontinuing due to MECs (1%). The first event resolved in most patients (grade ≥2 MECs and visual acuity [each 77%], blurred vision [67%], and dry eye [86%]), with no reports of permanent vision loss to date. A literature review confirmed that similar MECs were reported with other ADCs; however, event management strategies varied. The pathophysiology of MECs is unclear, though the ADC cytotoxic payload may contribute to on- or off-target effects on corneal epithelial cells. CONCLUSION Single-agent belamaf represents a new treatment option for patients with RRMM. As with other ADCs, MECs were observed and additional research is warranted to determine their pathophysiology. A multidisciplinary approach, involving close collaboration between eye care professionals and hematologist/oncologists, is needed to determine appropriate diagnosis and management of these patients. TRIAL REGISTRATION ClinicalTrials.gov Identifier, NCT03525678.
Collapse
Affiliation(s)
- Asim V Farooq
- University of Chicago Medical Center, Chicago, IL, USA.
| | - Simona Degli Esposti
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust, UCL Institute of Ophthalmology, London, UK
| | - Rakesh Popat
- University College London Hospitals, NHS Foundation Trust, London, UK
| | | | - Sagar Lonial
- Emory University, Winship Cancer Institute, Atlanta, GA, USA
| | - Ajay K Nooka
- Emory University, Winship Cancer Institute, Atlanta, GA, USA
| | | | - Douglas Sborov
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Brian E Zaugg
- Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Ashraf Z Badros
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bennie H Jeng
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Ira Gupta
- GlaxoSmithKline, Upper Providence, PA, USA
| | - Kathryn Colby
- University of Chicago Medical Center, Chicago, IL, USA
| |
Collapse
|
28
|
Kopylov AM, Zavyalova EG, Pavlova GV, Pronin IN. [Theranostics for glioblastoma with monoclonal antibodies to the epidermal growth factor receptor]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2020; 84:113-118. [PMID: 32649821 DOI: 10.17116/neiro202084031113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A review is devoted to analysis of the prospects of theranostics for multiform glioblastoma with monoclonal antibodies to the epidermal growth factor receptor (EGFR). Treatment of various malignancies demonstrated high potential of the use of EGFR. However, in case of glioblastoma, the effectiveness of monoclonal antibodies to EGFR is constrained by the absence of informative criteria for assessing the effectiveness of diagnosis and treatment of disease.
Collapse
Affiliation(s)
- A M Kopylov
- Lomonosov Moscow State University, Moscow, Russia.,Apto-Pharm LLC, Moscow, Russia
| | - E G Zavyalova
- Lomonosov Moscow State University, Moscow, Russia.,Apto-Pharm LLC, Moscow, Russia
| | - G V Pavlova
- Apto-Pharm LLC, Moscow, Russia.,Institute of Gene Biology of RAS, Moscow, Russia.,Burdenko Neurosurgical Center, Moscow, Russia
| | - I N Pronin
- Burdenko Neurosurgical Center, Moscow, Russia
| |
Collapse
|
29
|
Lakomy R, Kazda T, Selingerova I, Poprach A, Pospisil P, Belanova R, Fadrus P, Vybihal V, Smrcka M, Jancalek R, Hynkova L, Muckova K, Hendrych M, Sana J, Slaby O, Slampa P. Real-World Evidence in Glioblastoma: Stupp's Regimen After a Decade. Front Oncol 2020; 10:840. [PMID: 32719739 PMCID: PMC7348058 DOI: 10.3389/fonc.2020.00840] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/28/2020] [Indexed: 11/13/2022] Open
Abstract
The aim of this retrospective study is to provide real-world evidence in glioblastoma treatment and to compare overall survival after Stupp's regimen treatment today and a decade ago. A current consecutive cohort of histologically confirmed glioblastoma irradiated from 1/2014 to 12/2017 in our cancer center was compared with an already published historical control of patients treated in 1/2003-12/2009. A total of new 155 patients was analyzed, median age 60.9 years, 61% men, 58 patients (37%) underwent gross total tumor resection. Stupp's regimen was indicated in 90 patients (58%), 65 patients (42%) underwent radiotherapy alone. Median progression-free survival in Stupp's regimen cohort was 6.7 months, median OS 16.0 months, and 2-year OS 30.7%. OS was longer if patients were able to finish at least three cycles of adjuvant chemotherapy (median 23.3 months and 43.9% of patients lived at 2 years after surgery). Rapid early progression prior to radiotherapy was a negative prognostic factor with HR 1.87 (p = 0.007). The interval between surgery and the start of radiotherapy (median 6.7 weeks) was not prognostically significant (p = 0.825). The median OS in the current cohort was about 2 months longer than in the historical control group treated 10 years ago (16 vs. 13.8 months) using the same Stupp's regimen. Taking into account differences in patient's characteristics between current and historical cohorts, age, extent of resection, and ECOG patient performance status adjusted HR (Stupp's regimen vs. RT alone) for OS was determined as 0.45 (p = 0.002).
Collapse
Affiliation(s)
- Radek Lakomy
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Tomas Kazda
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Research Center for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Iveta Selingerova
- Research Center for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Alexandr Poprach
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petr Pospisil
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Renata Belanova
- Department of Radiology, Masaryk Memorial Cancer Institute, Brno, Czechia.,Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Pavel Fadrus
- Department of Neurosurgery, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Vaclav Vybihal
- Department of Neurosurgery, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Radim Jancalek
- Department of Neurosurgery, St. Anne's University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Ludmila Hynkova
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katarina Muckova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Michal Hendrych
- First Department of Pathology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jiri Sana
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Ondrej Slaby
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Pavel Slampa
- Department of Radiation Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia.,Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
30
|
Parrozzani R, Lombardi G, Midena E, Leonardi F, Londei D, Padovan M, Caccese M, Marchione G, Bini S, Zagonel V, Frizziero L. Corneal side effects induced by EGFR-inhibitor antibody-drug conjugate ABT-414 in patients with recurrent glioblastoma: a prospective clinical and confocal microscopy study. Ther Adv Med Oncol 2020; 12:1758835920907543. [PMID: 32550861 PMCID: PMC7278095 DOI: 10.1177/1758835920907543] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/20/2020] [Indexed: 11/17/2022] Open
Abstract
Background: The aim of this study was to prospectively analyse, for the first time
worldwide by in vivo clinical confocal microscopy (CCM),
corneal side effects secondary to the use of epidermal growth factor
receptor (EGFR) inhibitor depatuxizumab mafodotin (ABT-414) in a cohort of
patients affected by EGFR-amplified recurrent glioblastoma. Methods: Each enrolled patient underwent full ophthalmologic examination including
in vivo CCM of the cornea. Each patient was examined at
baseline and every 2 weeks during treatment as long as patient conditions
allowed it. Results: A total of 10 patients were consecutively enrolled. Median follow-up was
5 months. No Common Terminology Criteria for Adverse Events Version 4.0
grade 4 toxicity was documented. Two (20%) grade 3 toxicities were
documented at week 8. CCM examination detected in all eyes multiple and
diffuse hyperreflective white round spots in the corneal basal epithelial
layers (100%), progressive subbasal nerve plexus layer fibres fragmentation
followed by full disappearance (100%) and appearance of round cystic
structures in the corneal epithelium (100%). All CCM documented side effects
reached the peak of prevalence and severity after a median of 3 infusions.
After treatment discontinuation, the reversibility of corneal side effects
was documented at CCM after a median of 4 weeks. Conclusion: ABT-414 toxicity is not only directed to the corneal epithelium, but also to
corneal nerves. Side effects are detectable in all treated patients and CCM
documents early corneal epithelium and subbasal nerve plexus toxicity, with
subsequent progressive restoration after treatment discontinuation. Ocular
side effects due to ABT-414 can be manageable.
Collapse
Affiliation(s)
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Edoardo Midena
- Department of Ophthalmology, University of Padova, Via Giustiniani 2, Padova, 35128, Italy IRCCS - Fondazione Bietti, Rome, Italy
| | | | - Davide Londei
- Department of Ophthalmology, University of Padova, Padova, Italy
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Giulia Marchione
- Department of Ophthalmology, University of Padova, Padova, Italy
| | - Silvia Bini
- Department of Ophthalmology, University of Padova, Padova, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | |
Collapse
|
31
|
Lobbous M, Nabors LB. A troublesome burden, the amplification of EGFR in glioblastoma! Neuro Oncol 2020; 22:594-595. [PMID: 32144420 PMCID: PMC7229254 DOI: 10.1093/neuonc/noaa055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Mina Lobbous
- Division of Neuro-oncology, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama
| | - L Burt Nabors
- Division of Neuro-oncology, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
32
|
Lee A, Arasaratnam M, Chan DLH, Khasraw M, Howell VM, Wheeler H. Anti-epidermal growth factor receptor therapy for glioblastoma in adults. Cochrane Database Syst Rev 2020; 5:CD013238. [PMID: 32395825 PMCID: PMC7389448 DOI: 10.1002/14651858.cd013238.pub2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Glioblastoma is an uncommon but highly aggressive type of brain tumour. Significant gains have been achieved in the molecular understanding and the pathogenesis of glioblastomas, however clinical improvements are difficult to obtain for many reasons. The current standard of care involves maximal safe surgical resection followed by chemoradiation and then adjuvant chemotherapy European Organisation for Research and Treatment of Cancer and the NCIC Clinical Trials Group (EORTC-NCIC) protocol with a median survival of 14.6 months. Successive phase III international randomised controlled studies have failed to significantly demonstrate survival advantage with newer drugs. Epidermal growth factor receptor (EGFR) is observed to be aberrant in 30% to 60% of glioblastomas. The receptor aberrancy is driven by abnormal gene amplification, receptor mutation, or both, in particular the extracellular vIII domain. EGFR abnormalities are common in solid tumours, and the advent of anti-EGFR therapies in non-small cell lung cancer and colorectal adenocarcinomas have greatly improved clinical outcomes. Anti-EGFR therapies have been investigated amongst glioblastomas, however questions remain about its ongoing role in glioblastoma management. This review aimed to report on the available evidence to date and perform a systematic analysis on the risks and benefits of use of anti-EGFR therapies in glioblastomas. OBJECTIVES To evaluate the efficacy and harms of anti-EGFR therapies for glioblastoma in adults. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, EBM Reviews databases, with supplementary handsearches to identify all available and relevant studies to 20 April 2020. SELECTION CRITERIA All randomised controlled trials (RCTs) using anti-EGFR therapies in adults with glioblastoma were eligible for inclusion. Anti-EGFR therapies included tyrosine kinase inhibitors, monoclonal antibodies, or vaccines. The comparison included investigational product added to standard of care versus standard of care or placebo, or investigational product against standard of care or placebo. DATA COLLECTION AND ANALYSIS The authorship team screened the search results and recorded the extracted data for analysis. We used standard Cochrane methodology to performed quantitative meta-analysis if two or more studies had appropriate and available data. Otherwise, we conducted a qualitative and descriptive analysis. We used the GRADE system to rate the certainty of the evidence. The analysis was performed along the two clinical settings: first-line (after surgery) and recurrent disease (after failure of first line treatment). Where information was available, we documented overall survival, progression-free survival, adverse events, and quality of life data from eligible studies. MAIN RESULTS The combined searches initially identified 912 records (after removal of duplicates), and further screening resulted in 19 records for full consideration. We identified nine eligible studies for inclusion in the review. There were three first-line studies and six recurrent studies. Five studies used tyrosine kinase inhibitors (TKIs); two studies used monoclonal antibodies; and two studies used targeted vaccines. More recent studies presented greater detail in the conduct of their studies and thus had a lower risk of bias. We observed no evidence benefit in overall survival with the use of anti-EGFR therapy in the first-line or recurrent setting (hazard ratio (HR) 0.89, 95% confidence interval (CI) 0.76 to 1.04; 3 RCTs, 1000 participants, moderate-certainty evidence; and HR 0.79, 95% CI 0.51 to 1.21, 4 RCTs, 489 participants, low-certainty evidence, respectively). All the interventions were generally well tolerated with low-certainty evidence for lymphopenia (odds ratio (OR) 0.97, 95% CI 0.19 to 4.81; 4 RCTs, 1146 participants), neutropenia (OR 1.29, 95% CI 0.82 to 2.03; 4 RCTs, 1146 participants), and thrombocytopenia (OR 3.69, 95% CI 0.51 to 26.51; 4 RCTs, 1146 participants). A notable toxicity relates to ABT-414, where significant ocular issues were detected. The addition of anti-EGFR therapy showed no evidence of an increase in progression-free survival (PFS) in the first-line setting (HR 0.94, 95% CI 0.81 to 1.10; 2 RCTs, 894 participants, low-certainty evidence). In the recurrent setting, there was an increase in PFS with the use of anti-EGFR therapy (HR 0.75, 95% CI 0.58 to 0.96, 3 RCTs, 275 participants, low-certainty evidence). The available quality of life assessment data showed that anti-EGFR therapies were neither detrimental or beneficial when compared to standard care (not estimable). AUTHORS' CONCLUSIONS In summary, there is no evidence of a demonstrable overall survival benefit with the addition of anti-EGFR therapy in first-line and recurrent glioblastomas. Newer drugs that are specially designed for glioblastoma targets may raise the possibility of success in this population, but data are lacking at present. Future studies should be more selective in pursuing people displaying specific EGFR targets.
Collapse
Affiliation(s)
- Adrian Lee
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | | | - David Lok Hang Chan
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, The University of Sydney, Camperdown, Australia
| | - Viive M Howell
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Helen Wheeler
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
33
|
Struve N, Binder ZA, Stead LF, Brend T, Bagley SJ, Faulkner C, Ott L, Müller-Goebel J, Weik AS, Hoffer K, Krug L, Rieckmann T, Bußmann L, Henze M, Morrissette JJD, Kurian KM, Schüller U, Petersen C, Rothkamm K, O Rourke DM, Short SC, Kriegs M. EGFRvIII upregulates DNA mismatch repair resulting in increased temozolomide sensitivity of MGMT promoter methylated glioblastoma. Oncogene 2020; 39:3041-3055. [PMID: 32066879 PMCID: PMC7142016 DOI: 10.1038/s41388-020-1208-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 11/08/2022]
Abstract
The oncogene epidermal growth factor receptor variant III (EGFRvIII) is frequently expressed in glioblastomas (GBM) but its impact on therapy response is still under controversial debate. Here we wanted to test if EGFRvIII influences the sensitivity towards the alkylating agent temozolomide (TMZ). Therefore, we retrospectively analyzed the survival of 336 GBM patients, demonstrating that under standard treatment, which includes TMZ, EGFRvIII expression is associated with prolonged survival, but only in patients with O6-methylguanine-DNA methyltransferase (MGMT) promoter methylated tumors. Using isogenic GBM cell lines with endogenous EGFRvIII expression we could demonstrate that EGFRvIII increases TMZ sensitivity and results in enhanced numbers of DNA double-strand breaks and a pronounced S/G2-phase arrest after TMZ treatment. We observed a higher expression of DNA mismatch repair (MMR) proteins in EGFRvIII+ cells and patient tumor samples, which was most pronounced for MSH2 and MSH6. EGFRvIII-specific knockdown reduced MMR protein expression thereby increasing TMZ resistance. Subsequent functional kinome profiling revealed an increased activation of p38- and ERK1/2-dependent signaling in EGFRvIII expressing cells, which regulates MMR protein expression downstream of EGFRvIII. In summary, our results demonstrate that the oncoprotein EGFRvIII sensitizes a fraction of GBM to current standard of care treatment through the upregulation of DNA MMR.
Collapse
Affiliation(s)
- Nina Struve
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Lucy F Stead
- Leeds Institute of Medical Research at St James's, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, UK
| | - Tim Brend
- Leeds Institute of Medical Research at St James's, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, UK
| | - Stephen J Bagley
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Claire Faulkner
- Bristol Genetics Laboratory, Southmead Hospital, Bristol, UK
| | - Leonie Ott
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Justus Müller-Goebel
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna-Sophie Weik
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstantin Hoffer
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leonie Krug
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Thorsten Rieckmann
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Lara Bußmann
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Marvin Henze
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kathreena M Kurian
- Bristol Brain Tumour Research Centre, University of Bristol, Bristol, UK
| | - Ulrich Schüller
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Petersen
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Radiotherapy and Radiooncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Donald M O Rourke
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Susan C Short
- Leeds Institute of Medical Research at St James's, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, UK
| | - Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiation Oncology, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
A phase 1 study evaluating safety and pharmacokinetics of losatuxizumab vedotin (ABBV-221), an anti-EGFR antibody-drug conjugate carrying monomethyl auristatin E, in patients with solid tumors likely to overexpress EGFR. Invest New Drugs 2020; 38:1483-1494. [PMID: 32189093 DOI: 10.1007/s10637-020-00908-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/30/2020] [Indexed: 01/25/2023]
Abstract
Losatuxizumab vedotin (formerly ABBV-221) is a second-generation antibody-drug conjugate targeting epidermal growth factor receptor (EGFR). In this multicenter phase 1 study, eligible patients with EGFR-dependent solid tumors received losatuxizumab vedotin (3 + 3 design) intravenously at starting dose of 0.3 mg/kg over 3 h per 21-day cycle, with alternate dosing schedules utilized (2 weeks on/1 week off or weekly) to mitigate infusion reactions. Forty-five patients received ≥1 doses of losatuxizumab vedotin (13 colon, 6 non-small cell lung cancer, 5 head and neck [HNC], 5 glioblastoma multiforme, 2 breast, 14 other). Tumor samples were evaluated for EGFR protein expression by immunohistochemistry, EGFR and EGFR ligand mRNA expression by RNAseq, and results compared with outcome. Most common adverse events were infusion-related reaction (22/45; 49%) and fatigue (20/45; 44%). While most infusion reactions were grade ≤ 2, four patients experienced grade ≥3 infusion reactions. Several infusion reaction mitigation strategies were explored. Because of the high incidence of infusion reactions, the trial was stopped and the maximum tolerated dose was not reached. The last cleared dose: 6 mg/kg/cycle. Nineteen patients (42%) had stable disease; 4 remained on study >6 months. One HNC patient with increased levels of EGFR and EGFR ligands (amphiregulin, epiregulin) achieved a confirmed partial response. Pharmacokinetic analysis of losatuxizumab vedotin showed exposures appeared to be approximately dose-proportional. The high frequency of infusion reactions necessitated early closure of this trial. The detailed mitigation strategies used in this protocol for infusion-related reactions may provide beneficial information for trial design of agents with high infusion reaction rates.
Collapse
|
35
|
Popat R, Warcel D, O'Nions J, Cowley A, Smith S, Tucker WR, Yong K, Esposti SD. Characterization of response and corneal events with extended follow-up after belantamab mafodotin (GSK2857916) monotherapy for patients with relapsed multiple myeloma: a case series from the first-time-in-human clinical trial. Haematologica 2020; 105:e261-e263. [PMID: 32107339 DOI: 10.3324/haematol.2019.235937] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Rakesh Popat
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Dana Warcel
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Jenny O'Nions
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Anna Cowley
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Sasha Smith
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - William R Tucker
- National Institute for Health Research Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital NHS Foundation Trust & University College London Institute of Ophthalmology, London, UK
| | - Kwee Yong
- National Institute for Health Research Clinical Research Facility, University College London Hospitals NHS Foundation Trust
| | - Simona Degli Esposti
- National Institute for Health Research Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital NHS Foundation Trust & University College London Institute of Ophthalmology, London, UK
| |
Collapse
|
36
|
Salvati M, Bruzzaniti P, Relucenti M, Nizzola M, Familiari P, Giugliano M, Scafa AK, Galletta S, Li X, Chen R, Barbaranelli C, Frati A, Santoro A. Retrospective and Randomized Analysis of Influence and Correlation of Clinical and Molecular Prognostic Factors in a Mono-Operative Series of 122 Patients with Glioblastoma Treated with STR or GTR. Brain Sci 2020; 10:brainsci10020091. [PMID: 32050461 PMCID: PMC7071604 DOI: 10.3390/brainsci10020091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma is a solid, infiltrating, and the most frequent highly malignant primary brain tumor. Our aim was to find the correlation between sex, age, preoperative Karnofsky performance status (KPS), presenting with seizures, and extent of resection (EOR) with overall survival (OS), progression-free survival (PFS), and postoperative KPS, along with the prognostic value of IDH1, MGMT, ATRX, EGFR, and TP53 genes mutations and of Ki67 through the analysis of a single-operator series in order to avoid the biases of a multi-operator series, such as the lack of homogeneity in surgical and adjuvant nonsurgical treatments. A randomized retrospective analysis of 122 patients treated by a single first operator at Sapienza University of Rome was carried out. After surgery, patients followed standard Stupp protocol treatment. Exclusion criteria were: (1) patients with primary brainstem and spinal cord gliomas and (2) patients who underwent partial resections (resection < 90%) or a biopsy exclusively for diagnostic purposes. Statistical analysis with a simultaneous regression model was carried out through the use of SPSS 25® (IBM). Results showed statistically significant survival increase in four groups: (1) patients treated with gross total resection (GTR) (p < 0.030); (2) patients with mutation of IDH1 (p < 0.0161); (3) patients with methylated MGMT promoter (p < 0.005); (4) patients without EGFR amplification or EGFRvIII mutation (p < 0.035). Higher but not statistically significant survival rates were also observed in: patients <75 years, patients presenting with seizures at diagnosis, patients affected by lesions in noneloquent areas, as well as in patients with ATRX gene mutation and Ki-67 < 10%.
Collapse
Affiliation(s)
- Maurizio Salvati
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Placido Bruzzaniti
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
- Correspondence: ; Tel.: +39-3349753520
| | - Michela Relucenti
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Science, “La Sapienza” University of Rome, 00161 Rome, Italy;
| | - Mariagrazia Nizzola
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Pietro Familiari
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Marco Giugliano
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Anthony Kevin Scafa
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| | - Santi Galletta
- UOSD of Neurophysiopathology and DISMOV, AOU G Martino, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing 210009, China; (X.L.); (R.C.)
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Dingjiaqiao 87, Nanjing 210009, China; (X.L.); (R.C.)
| | - Claudio Barbaranelli
- Department of Psychology, Faculty of Medicine and Psychology “La Sapienza” University of Rome, 00189 Rome, Italy;
| | - Alessandro Frati
- Department of Neurosurgery, IRCCS Neuromed Pozzilli IS, 86077 Isernia, Italy;
| | - Antonio Santoro
- Department of Neurological Sciences, Neurosurgey, “La Sapienza” University of Rome, 00161 Rome, Italy; (M.S.); (M.N.); (P.F.); (M.G.); (A.K.S.); (A.S.)
| |
Collapse
|
37
|
Shin E, Lim DH, Han J, Nam DH, Park K, Ahn MJ, Kang WK, Lee J, Ahn JS, Lee SH, Sun JM, Jung HA, Chung TY. Markedly increased ocular side effect causing severe vision deterioration after chemotherapy using new or investigational epidermal or fibroblast growth factor receptor inhibitors. BMC Ophthalmol 2020; 20:19. [PMID: 31918686 PMCID: PMC6953164 DOI: 10.1186/s12886-019-1285-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022] Open
Abstract
Background We sought to describe corneal epithelial changes after using epidermal (EGFR) or fibroblast growth factor receptor (FGFR) inhibitors as chemotherapy and to clarify incidence and prognosis. Materials Retrospective chart review. Results Among 6871 patients and 17 EGFR or FGFR inhibitors, 1161 patients (16.9%) referred for ophthalmologic examination. In total, 1145 patients had disease-related or unrelated ocular complications. Among 16 patients with treatment-related ocular complications, three patients had treatment-related radiation retinopathy and one patient showed treatment-related corneal ulcer. Finally the authors identified that, in 12 patients, three EGFR inhibitors and two FGFR inhibitors caused corneal epithelial lesions. Vandetanib, Osimertinib, and ABT-414 caused vortex keratopathy in nine patients, while ASP-5878 and FPA-144 caused epithelial changes resembling corneal dysmaturation in three patients. The mean interval until symptoms appeared was 246 days with vandetanib, 196 days with osimertinib, 30 days with ABT-414, 55 days with ASP-5878, and 70 days with FPA-144. The mean of the lowest logarithm of minimal angle of resolution visual acuity results of the right and left eyes after chemotherapy were 0.338 and 0.413. The incidence rates of epithelial changes were 15.79% with vandetanib, 0.5% with osimertinib, 100% with ABT-414, 50.0% with ASP-5878, and 18.2% with FPA-144. After excluding deceased patients and those who were lost to follow-up or still undergoing treatment, we confirmed the reversibility of corneal lesions after the discontinuation of each agent. Seven patients showed full recovery of their vision and corneal epithelium, while three achieved a partial level of recovery. Although patients diagnosed with glioblastoma used prophylactic topical steroids before and during ABT-414 therapy, all developed vortex keratopathy. Conclusions EGFR and FGFR inhibitors are chemotherapy agents that could make corneal epithelial changes. Contrary to the low probability of ocular complication with old EGFR drugs, recently introduced EGFR and FGFR agents showed a high incidence of ocular complication with severe vision distortion. Doctors should forewarn patients planning chemotherapy with these agents that decreased visual acuity could develop due to corneal epithelial changes and also reassure them that the condition could be improved after the end of treatment without the use of steroid eye drops. Trial registration This study was approved by the institutional review board (IRB) of Samsung Medical Center (IRB no. 2019–04-027) and was conducted according to the principles expressed in the Declaration of Helsinki.
Collapse
Affiliation(s)
- Eunhae Shin
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Hui Lim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea. .,Department of Preventive Medicine, Graduate School, Catholic University of Korea, Seoul, Republic of Korea.
| | - Jisang Han
- Department of Ophthalmology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Do-Hyun Nam
- Cancer Stem Cell Research Center, Department of Neurosurgery, Samsung Medical Center and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tae-Young Chung
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Abdelaziz MO, Ossmann S, Kaufmann AM, Leitner J, Steinberger P, Willimsky G, Raftery MJ, Schönrich G. Development of a Human Cytomegalovirus (HCMV)-Based Therapeutic Cancer Vaccine Uncovers a Previously Unsuspected Viral Block of MHC Class I Antigen Presentation. Front Immunol 2019; 10:1776. [PMID: 31417555 PMCID: PMC6682651 DOI: 10.3389/fimmu.2019.01776] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) induces a uniquely high frequency of virus-specific effector/memory CD8+ T-cells, a phenomenon termed “memory inflation”. Thus, HCMV-based vaccines are particularly interesting in order to stimulate a sustained and strong cellular immune response against cancer. Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with high lethality and inevitable relapse. The current standard treatment does not significantly improve the desperate situation underlining the urgent need to develop novel approaches. Although HCMV is highly fastidious with regard to species and cell type, GBM cell lines are susceptible to HCMV. In order to generate HCMV-based therapeutic vaccine candidates, we deleted all HCMV-encoded proteins (immunoevasins) that interfere with MHC class I presentation. The aim being to use the viral vector as an adjuvant for presentation of endogenous tumor antigens, the presentation of high levels of vector-encoded neoantigens and finally the repurposing of bystander HCMV-specific CD8+ T cells to fight the tumor. As neoantigen, we exemplarily used the E6 and E7 proteins of human papillomavirus type 16 (HPV-16) as a non-transforming fusion protein (E6/E7) that covers all relevant antigenic peptides. Surprisingly, GBM cells infected with E6/E7-expressing HCMV-vectors failed to stimulate E6-specific T cells despite high level expression of E6/E7 protein. Further experiments revealed that MHC class I presentation of E6/E7 is impaired by the HCMV-vector although it lacks all known immunoevasins. We also generated HCMV-based vectors that express E6-derived peptide fused to HCMV proteins. GBM cells infected with these vectors efficiently stimulated E6-specific T cells. Thus, fusion of antigenic sequences to HCMV proteins is required for efficient presentation via MHC class I molecules during infection. Taken together, these results provide the preclinical basis for development of HCMV-based vaccines and also reveal a novel HCMV-encoded block of MHC class I presentation.
Collapse
Affiliation(s)
- Mohammed O Abdelaziz
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sophia Ossmann
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Andreas M Kaufmann
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, Partner Site Berlin, Berlin, Germany
| | - Martin J Raftery
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Günther Schönrich
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
39
|
El‐Deiry WS, Goldberg RM, Lenz H, Shields AF, Gibney GT, Tan AR, Brown J, Eisenberg B, Heath EI, Phuphanich S, Kim E, Brenner AJ, Marshall JL. The current state of molecular testing in the treatment of patients with solid tumors, 2019. CA Cancer J Clin 2019; 69:305-343. [PMID: 31116423 PMCID: PMC6767457 DOI: 10.3322/caac.21560] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The world of molecular profiling has undergone revolutionary changes over the last few years as knowledge, technology, and even standard clinical practice have evolved. Broad molecular profiling is now nearly essential for all patients with metastatic solid tumors. New agents have been approved based on molecular testing instead of tumor site of origin. Molecular profiling methodologies have likewise changed such that tests that were performed on patients a few years ago are no longer complete and possibly inaccurate today. As with all rapid change, medical providers can quickly fall behind or struggle to find up-to-date sources to ensure he or she provides optimum care. In this review, the authors provide the current state of the art for molecular profiling/precision medicine, practice standards, and a view into the future ahead.
Collapse
Affiliation(s)
- Wafik S. El‐Deiry
- Associate Dean for Oncologic Sciences, Warren Alpert Medical School; Director, Joint Program in Cancer Biology, Brown University and the Lifespan Cancer Institute; Professor of Pathology & Laboratory Medicine and Professor of Medical ScienceBrown UniversityProvidenceRI
| | - Richard M. Goldberg
- Professor of Medicine and DirectorWest Virginia University Cancer InstituteMorgantownWV
| | - Heinz‐Josef Lenz
- Professor of Medicine, Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCA
| | | | - Geoffrey T. Gibney
- Associate Professor of Medicine, Co‐Leader of the Melanoma Disease GroupLombardi Comprehensive Cancer Institute, MedStar Georgetown Cancer InstituteWashingtonDC
| | - Antoinette R. Tan
- Co‐Director of Phase I Program, Department of Solid Tumor Oncology and Investigational TherapeuticsLevine Cancer Institute, Atrium HealthCharlotteNC
| | - Jubilee Brown
- Professor and Associate Director of Gynecologic OncologyLevine Cancer Institute, Atrium HealthCharlotteNC
| | - Burton Eisenberg
- Professor of Clinical SurgeryUniversity of Southern CaliforniaLos AngelesCA
- Executive Medical DirectorHoag Family Cancer InstituteNewport BeachCA
| | | | - Surasak Phuphanich
- Professor of Neurology, Director, Division of Neuro‐OncologyBarrow Neurological InstitutePhoenixAZ
| | - Edward Kim
- Chair, Solid Tumor Oncology and Investigational TherapeuticsLevine Cancer Institute, Atrium HealthCharlotteNC
| | - Andrew J. Brenner
- Associate Professor of Medicine, Mays Cancer Center at University of Texas Health San Antonio Cancer CenterSan AntonioTX
| | - John L. Marshall
- Professor of Medicine and Oncology, Director, Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer InstituteMedStar Georgetown Cancer InstituteWashingtonDC
| |
Collapse
|
40
|
Lassman AB, Roberts-Rapp L, Sokolova I, Song M, Pestova E, Kular R, Mullen C, Zha Z, Lu X, Gomez E, Bhathena A, Maag D, Kumthekar P, Gan HK, Scott AM, Guseva M, Holen KD, Ansell PJ, van den Bent MJ. Comparison of Biomarker Assays for EGFR: Implications for Precision Medicine in Patients with Glioblastoma. Clin Cancer Res 2019; 25:3259-3265. [PMID: 30796037 PMCID: PMC8291723 DOI: 10.1158/1078-0432.ccr-18-3034] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/16/2019] [Accepted: 02/18/2019] [Indexed: 12/26/2022]
Abstract
PURPOSE Patients with glioblastoma (GBM) have a poor prognosis and are in desperate need of better therapies. As therapeutic decisions are increasingly guided by biomarkers, and EGFR abnormalities are common in GBM, thus representing a potential therapeutic target, we systematically evaluated methods of assessing EGFR amplification by multiple assays. Specifically, we evaluated correlation among fluorescence in situ hybridization (FISH), a standard assay for detecting EGFR amplification, with other methods.Experimental Design: Formalin-fixed, paraffin-embedded tumor samples were used for all assays. EGFR amplification was detected using FISH (N = 206) and whole-exome sequencing (WES, N = 74). EGFR mRNA expression was measured using reverse transcription-polymerase chain reaction (RT-PCR, N = 206) and transcriptome profiling (RNAseq, N = 64). EGFR protein expression was determined by immunohistochemistry (IHC, N = 34). Significant correlations among various methods were determined using Cohen's kappa (κ = 0.61-0.80 defines substantial agreement) or R 2 statistics. RESULTS EGFR mRNA expression levels by RNA sequencing (RNAseq) and RT-PCR were highly correlated with EGFR amplification assessed by FISH (κ = 0.702). High concordance was also observed when comparing FISH to WES (κ = 0.739). RNA expression was superior to protein expression in delineating EGFR amplification. CONCLUSIONS Methods for assessing EGFR mRNA expression (RT-PCR, RNAseq) and copy number (WES), but not protein expression (IHC), can be used as surrogates for EGFR amplification (FISH) in GBM. Collectively, our results provide enhanced understanding of available screening options for patients, which may help guide EGFR-targeted therapeutic approaches.
Collapse
Affiliation(s)
- Andrew B Lassman
- Department of Neurology & Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York.
| | | | | | | | | | | | | | - Zheng Zha
- AbbVie, Inc., North Chicago, Illinois
| | - Xin Lu
- AbbVie, Inc., North Chicago, Illinois
| | | | | | | | - Priya Kumthekar
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Hui K Gan
- Austin Health, Olivia Newton-John Cancer Research Institute, and La Trobe University, Melbourne, Victoria, Australia
| | - Andrew M Scott
- Austin Health, Olivia Newton-John Cancer Research Institute, and La Trobe University, Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
41
|
Nasrallah MP, Binder ZA, Oldridge DA, Zhao J, Lieberman DB, Roth JJ, Watt CD, Sukhadia S, Klinman E, Daber RD, Desai A, Brem S, O'Rourke DM, Morrissette JJD. Molecular Neuropathology in Practice: Clinical Profiling and Integrative Analysis of Molecular Alterations in Glioblastoma. Acad Pathol 2019; 6:2374289519848353. [PMID: 31206012 PMCID: PMC6537274 DOI: 10.1177/2374289519848353] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 02/27/2019] [Accepted: 03/25/2019] [Indexed: 12/20/2022] Open
Abstract
Molecular profiling of glioblastoma has revealed complex cytogenetic, epigenetic, and molecular abnormalities that are necessary for diagnosis, prognosis, and treatment. Our neuro-oncology group has developed a data-driven, institutional consensus guideline for efficient and optimal workup of glioblastomas based on our routine performance of molecular testing. We describe our institution’s testing algorithm, assay development, and genetic findings in glioblastoma, to illustrate current practices and challenges in neuropathology related to molecular and genetic testing. We have found that coordination of test requisition, tissue handling, and incorporation of results into the final pathologic diagnosis by the neuropathologist improve patient care. Here, we present analysis of O6-methylguanine-DNA-methyltransferase promoter methylation and next-generation sequencing results of 189 patients, obtained utilizing our internal processes led by the neuropathology team. Our institutional pathway for neuropathologist-driven molecular testing has streamlined the management of glioblastoma samples for efficient return of results for incorporation of genomic data into the pathological diagnosis and optimal patient care.
Collapse
Affiliation(s)
- MacLean P Nasrallah
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Derek A Oldridge
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianhua Zhao
- Bioreference Laboratories, West Deptford, NJ, USA
| | - David B Lieberman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacquelyn J Roth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher D Watt
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shrey Sukhadia
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eva Klinman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Arati Desai
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Brem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
42
|
Bertero L, Siravegna G, Rudà R, Soffietti R, Bardelli A, Cassoni P. Review: Peering through a keyhole: liquid biopsy in primary and metastatic central nervous system tumours. Neuropathol Appl Neurobiol 2019; 45:655-670. [PMID: 30977933 PMCID: PMC6899864 DOI: 10.1111/nan.12553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
Abstract
Tumour molecular profiling by liquid biopsy is being investigated for a wide range of research and clinical purposes. The possibility of repeatedly interrogating the tumour profile using minimally invasive procedures is helping to understand spatial and temporal tumour heterogeneity, and to shed a light on mechanisms of resistance to targeted therapies. Moreover, this approach has been already implemented in clinical practice to address specific decisions regarding patients’ follow‐up and therapeutic management. For central nervous system (CNS) tumours, molecular profiling is particularly relevant for the proper characterization of primary neoplasms, while CNS metastases can significantly diverge from primary disease or extra‐CNS metastases, thus compelling a dedicated assessment. Based on these considerations, effective liquid biopsy tools for CNS tumours are highly warranted and a significant amount of data have been accrued over the last few years. These results have shown that liquid biopsy can provide clinically meaningful information about both primary and metastatic CNS tumours, but specific considerations must be taken into account, for example, when choosing the source of liquid biopsy. Nevertheless, this approach is especially attractive for CNS tumours, as repeated tumour sampling is not feasible. The aim of our review was to thoroughly report the state‐of‐the‐art regarding the opportunities and challenges posed by liquid biopsy in both primary and secondary CNS tumours.
Collapse
Affiliation(s)
- L Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Torino, Italy.,Pathology Unit, Città della Salute e della Scienza University Hospital, Turin, Torino, Italy
| | - G Siravegna
- Department of Oncology, University of Turin, Candiolo (Turin), Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo (Turin), Italy
| | - R Rudà
- Neuro-oncology Unit, Department of Neurosciences, University of Turin, Italy.,Neuro-oncology Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - R Soffietti
- Neuro-oncology Unit, Department of Neurosciences, University of Turin, Italy.,Neuro-oncology Unit, Città della Salute e della Scienza University Hospital, Turin, Italy
| | - A Bardelli
- Department of Oncology, University of Turin, Candiolo (Turin), Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo (Turin), Italy
| | - P Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, Torino, Italy.,Pathology Unit, Città della Salute e della Scienza University Hospital, Turin, Torino, Italy
| |
Collapse
|
43
|
Kerstetter-Fogle A, Shukla S, Wang C, Beiss V, Harris PLR, Sloan AE, Steinmetz NF. Plant Virus-Like Particle In Situ Vaccine for Intracranial Glioma Immunotherapy. Cancers (Basel) 2019; 11:cancers11040515. [PMID: 30974896 PMCID: PMC6521079 DOI: 10.3390/cancers11040515] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
Despite aggressive multi-modality treatment with surgery, radiation and chemotherapies, malignant glioma inevitably recurs and has dismal survival rates. Recent progress in immunotherapy has led to a resurgence of interest, and immunotherapies are being investigated for treatment of glioma. However, the unique brain anatomy and a highly immunosuppressive glioma microenvironment pose significant challenges to achieving efficacy. Thus, there is a critical need for assessment of next-generation immunotherapies for glioma. In this study, we have investigated the efficacy of the nanoparticle platform technology based on plant-derived Cowpea mosaic virus like particles (empty CPMV or eCPMV) to instigate a potent immune response against intracranial glioma. CPMV immunotherapy has been shown to efficiently reverse the immunosuppressive tumor microenvironments in pre-clinical murine models of dermal melanoma and metastatic melanoma, metastatic breast cancer, intraperitoneal ovarian cancer and in canine patients with oral melanoma. In the present study, we demonstrate that in situ administration of CPMV immunotherapy in the setting of glioma can effectively recruit unique subset of effector innate and adaptive immune cells to the brain parenchyma while reducing immune suppressive cellular population, leading to regression of intracranial glioma. The in situ CPMV nanoparticle vaccine offers a potent yet safe and localized immunotherapy for intracranial glioma.
Collapse
Affiliation(s)
- Amber Kerstetter-Fogle
- Department of Neurological Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Sourabh Shukla
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Chao Wang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Veronique Beiss
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Peggy L R Harris
- Department of Neurological Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Andrew E Sloan
- Department of Neurological Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
- University Hospitals-Cleveland Medical Center & the Seidman Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, CA 92093, USA.
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Bumbaca B, Li Z, Shah DK. Pharmacokinetics of protein and peptide conjugates. Drug Metab Pharmacokinet 2019; 34:42-54. [PMID: 30573392 PMCID: PMC6378135 DOI: 10.1016/j.dmpk.2018.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Protein and peptide conjugates have become an important component of therapeutic and diagnostic medicine. These conjugates are primarily designed to improve pharmacokinetics (PK) of those therapeutic or imaging agents, which do not possess optimal disposition characteristics. In this review we have summarized preclinical and clinical PK of diverse protein and peptide conjugates, and have showcased how different conjugation approaches are used to obtain the desired PK. We have classified the conjugates into peptide conjugates, non-targeted protein conjugates, and targeted protein conjugates, and have highlighted diagnostic and therapeutic applications of these conjugates. In general, peptide conjugates demonstrate very short half-life and rapid renal elimination, and they are mainly designed to achieve high contrast ratio for imaging agents or to deliver therapeutic agents at sites not reachable by bulky or non-targeted proteins. Conjugates made from non-targeted proteins like albumin are designed to increase the half-life of rapidly eliminating therapeutic or imaging agents, and improve their delivery to tissues like solid tumors and inflamed joints. Targeted protein conjugates are mainly developed from antibodies, antibody derivatives, or endogenous proteins, and they are designed to improve the contrast ratio of imaging agents or therapeutic index of therapeutic agents, by enhancing their delivery to the site-of-action.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA.
| |
Collapse
|
45
|
Eskilsson E, Røsland GV, Solecki G, Wang Q, Harter PN, Graziani G, Verhaak RGW, Winkler F, Bjerkvig R, Miletic H. EGFR heterogeneity and implications for therapeutic intervention in glioblastoma. Neuro Oncol 2019; 20:743-752. [PMID: 29040782 DOI: 10.1093/neuonc/nox191] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Patients with glioblastoma (GBM) have a universally poor prognosis and are in urgent need of effective treatment strategies. Recent advances in sequencing techniques unraveled the complete genomic landscape of GBMs and revealed profound heterogeneity of individual tumors even at the single cell level. Genomic profiling has detected epidermal growth factor receptor (EGFR) gene alterations in more than half of GBMs. Major genetic events include amplification and mutation of EGFR. Yet, treatment strategies targeting EGFR have thus far failed in clinical trials. In this review, we discuss the clonal and functional heterogeneity of EGFRs in GBM development and critically reassess the potential of EGFRs as therapeutic targets.
Collapse
Affiliation(s)
- Eskil Eskilsson
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Gro V Røsland
- Department of Biomedicine, University of Bergen, Norway
| | - Gergely Solecki
- Department of Neurooncology, University Hospital Heidelberg, Germany
| | - Qianghu Wang
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.,Department of Bioinformatics and Computational Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Patrick N Harter
- Edinger-Institute, Goethe-University Medical School, Frankfurt am Main, Germany
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Roel G W Verhaak
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.,Department of Bioinformatics and Computational Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.,The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Frank Winkler
- Department of Neurooncology, University Hospital Heidelberg, Germany
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Brain Tumor Research Center, University of Bergen, Norway.,Norlux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Brain Tumor Research Center, University of Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
46
|
Abstract
This paper explicates the impact of tumor capillary permeability for glioma World Health Organization (WHO) grades II to IV on brain-penetrant drug entry and distribution within the tumor and the brain adjacent to tumor (leading edge). In addition, we consider the distribution of non-brain penetrant drugs and how, in some cases, large-molecular-weight drugs might achieve good distribution into tumor and brain adjacent to tumor.
Collapse
Affiliation(s)
- Victor A Levin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Neurosurgery, UCSF Medical School, San Francisco, California
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
47
|
Lassman AB, van den Bent MJ, Gan HK, Reardon DA, Kumthekar P, Butowski N, Lwin Z, Mikkelsen T, Nabors LB, Papadopoulos KP, Penas-Prado M, Simes J, Wheeler H, Walbert T, Scott AM, Gomez E, Lee HJ, Roberts-Rapp L, Xiong H, Ansell PJ, Bain E, Holen KD, Maag D, Merrell R. Safety and efficacy of depatuxizumab mafodotin + temozolomide in patients with EGFR-amplified, recurrent glioblastoma: results from an international phase I multicenter trial. Neuro Oncol 2019; 21:106-114. [PMID: 29982805 PMCID: PMC6303422 DOI: 10.1093/neuonc/noy091] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Patients with glioblastoma (GBM) have a dismal prognosis. Nearly all will relapse with no clear standard of care for recurrent disease (rGBM). Approximately 50% of patients have tumors harboring epidermal growth factor receptor (EGFR) amplification. The antibody-drug conjugate depatuxizumab mafodotin (depatux-m) binds cells with EGFR amplification, is internalized, and releases a microtubule toxin, killing the cell. Here we report efficacy, safety and pharmacokinetics (PK) of depatux-m + temozolomide (TMZ) in patients with EGFR-amplified rGBM. Methods M12-356 (NCT01800695) was an open-label study encompassing patients with newly diagnosed or rGBM across 3 treatment arms. Results are reported for adults with EGFR-amplified, measurable rGBM who received depatux-m (0.5-1.5 mg/kg) on days 1 and 15, and TMZ (150-200 mg/m2) on days 1-5 in a 28-day cycle. Patients were bevacizumab and nitrosourea naïve. Results There were 60 patients, median age 56 years (range, 20-79). Fifty-nine patients previously received TMZ. Common adverse events (AEs) were blurred vision (63%), fatigue (38%), and photophobia (35%). Grades 3/4 AEs were split between ocular and non-ocular AEs, occurring in 22% of patients each. Systemic PK exposure of depatux-m was dose proportional. The objective response rate was 14.3%, the 6-month progression-free survival rate was 25.2%, and the 6-month overall survival rate was 69.1%. Conclusions Depatux-m + TMZ displayed an AE profile similar to what was described previously. Antitumor activity in this TMZ-refractory population was encouraging. Continued study of depatux-m in patients with EGFR-amplified, newly diagnosed, or recurrent GBM is ongoing in 2 global, randomized trials (NCT02573324, NCT02343406).
Collapse
Affiliation(s)
- Andrew B Lassman
- Department of Neurology and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Hui K Gan
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Austin Health and Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Nicholas Butowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Zarnie Lwin
- Department of Medical Oncology, University of Queensland School of Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
| | | | - Louis B Nabors
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Marta Penas-Prado
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Helen Wheeler
- Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | | | - Andrew M Scott
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
- Austin Health and Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | | | - Ho-Jin Lee
- AbbVie Inc., North Chicago, Illinois, USA
| | | | - Hao Xiong
- AbbVie Inc., North Chicago, Illinois, USA
| | | | - Earle Bain
- AbbVie Inc., North Chicago, Illinois, USA
| | | | - David Maag
- AbbVie Inc., North Chicago, Illinois, USA
| | - Ryan Merrell
- NorthShore University Health System, Evanston, Illinois, USA
| |
Collapse
|
48
|
Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies (Basel) 2019; 8:E3. [PMID: 31544809 PMCID: PMC6640706 DOI: 10.3390/antib8010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.
Collapse
Affiliation(s)
- Andrew T Lucas
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Ryan Robinson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Joseph A Piscitelli
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - Juan F Razo
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
49
|
Eshraghi H, Mantopoulos D, Suh LH, Zaldana F, Fine HF. Corneal Toxicity Induced by ABT-414 Therapy for Glioblastoma Multiforme: A Case Report. Case Rep Ophthalmol 2018; 9:479-483. [PMID: 30631275 PMCID: PMC6323377 DOI: 10.1159/000494713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/18/2018] [Indexed: 11/19/2022] Open
Abstract
Background To describe a case of corneal toxicity associated with the use of the epidermal growth factor receptor (EGFR) inhibitor ABT-414 in the treatment of glioblastoma multiforme. Case Presentation Case report of a 56-year-old male with glioblastoma multiforme who developed mild painless blurred vision after systemic treatment with the investigational EGFR inhibitor ABT-414. The patient had best corrected visual acuity of 20/60 right eye and 20/50 left eye. Ophthalmic examination revealed corneal toxicity with whorl-like opacities in the inferior interpalpebral cornea. The patient was treated with topical fluoromethalone and lifitegrast, and his ocular symptoms as well as the corneal findings improved. Conclusions The systemic use of EGFR inhibitor ABT-414 may be associated with corneal toxicity and the effects are reversible with treatment.
Collapse
Affiliation(s)
- Hamoon Eshraghi
- Department of Ophthalmology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Dimosthenis Mantopoulos
- Department of Ophthalmology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Leejee H Suh
- Department of Ophthalmology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Francisco Zaldana
- Department of Pathology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Howard F Fine
- Department of Ophthalmology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
50
|
Optimising Outcomes for Glioblastoma through Subspecialisation in a Regional Cancer Centre. Brain Sci 2018; 8:brainsci8100186. [PMID: 30326653 PMCID: PMC6210056 DOI: 10.3390/brainsci8100186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 12/25/2022] Open
Abstract
Delivery of highly sophisticated, and subspecialised, management protocols for glioblastoma in low volume rural and regional areas creates potential issues for equivalent quality of care. This study aims to demonstrate the impact on clinical quality indicators through the development of a novel model of care delivering an outsourced subspecialised neuro-oncology service in a regional centre compared with the large volume metropolitan centre. Three hundred and fifty-two patients with glioblastoma were managed under the European Organisation for Research and Treatment of Cancer and National Cancer Institute of Canada Clinical Trials Group (EORTC-NCIC) Protocol, and survival outcome was assessed in relation to potential prognostic factors and the geographical site of treatment, before and after opening of a regional cancer centre. The median overall survival was 17 months (95% CI: 15.5–18.5), with more favourable outcome with age less than 50 years (p < 0.001), near-total resection (p < 0.001), Eastern Cooperative Oncology Group (ECOG) Performance status 0, 1 (p < 0.001), and presence of O-6 methylguanine DNA methyltransferase (MGMT) methylation (p = 0.001). There was no difference in survival outcome for patients managed at the regional centre, compared with metropolitan centre (p = 0.35). Similarly, no difference was seen with clinical quality process indicators of clinical trial involvement, rates of repeat craniotomy, use of bevacizumab and re-irradiation. This model of neuro-oncology subspecialisation allowed equivalent outcomes to be achieved within a regional cancer centre compared to large volume metropolitan centre.
Collapse
|