1
|
Millot F, Ampatzidou M, Moulik NR, Tewari S, Elhaddad A, Hammad M, Pichler H, Lion T, Tragiannidis A, Shima H, An W, Yang W, Karow A, Farah R, Luesink M, Dworzak M, Sembill S, De Moerloose B, Sedlacek P, Schultz KR, Kalwak K, Versluys B, Athale U, Hijiya N, Metzler M, Suttorp M. Management of children and adolescents with chronic myeloid leukemia in chronic phase: International pediatric chronic myeloid leukemia expert panel recommendations. Leukemia 2025; 39:779-791. [PMID: 40044960 DOI: 10.1038/s41375-025-02543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/30/2025] [Accepted: 02/13/2025] [Indexed: 04/09/2025]
Abstract
The treatment strategy for children and adolescents with chronic myeloid leukemia in the chronic phase (CML-CP) has evolved from allogeneic hematopoietic stem cell transplantation (HSCT) to tyrosine kinase inhibitors (TKIs). With the advent of next-generation TKIs and new targeted therapies in the CML field, an international pediatric CML expert panel provides recommendations based on the medical literature (including previous pediatric guidelines), national standards, and treatment principles used in adults with CML-CP. Recommendations include diagnosis of the disease and details on managing the initial steps of care of children and adolescents with newly diagnosed CML-CP, including complications such as leukostasis. The treatment recommendations are based on the initiation of therapy with a first- or second-generation TKI according to the allocated European Treatment and Outcome Study (EUTOS) long-term survival score risk group of the patient. The subsequent steps are based on the results of recommended monitoring which can justify a switch to another TKI or a drug in development if there is resistance or toxicity. The panel also provides recommendations regarding the discontinuation criteria for TKIs in children and adolescents in sustained deep molecular response. Allogeneic HSCT is not recommended as the first-line of treatment for children with CML-CP but is to be considered in case of progression to the advanced phase or failure of several lines of treatment. The present treatment and management recommendations are intended to provide advice to clinicians in view of optimizing the care and the outcome of children and adolescents with CML-CP.
Collapse
Affiliation(s)
- Frédéric Millot
- Inserm CIC 1402, University Hospital of Poitiers, Poitiers, France.
| | - Mirella Ampatzidou
- Department of Pediatric Hematology Oncology (T.A.O.), Aghia Sophia Children's Hospital, Athens, Greece
| | - Nirmalya Roy Moulik
- Department of Pediatric Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sanjay Tewari
- Department of Paediatric Oncology/Haematology, Royal Marsden NHS Foundation Trust Sutton, Sutton, UK
| | - Alaa Elhaddad
- Department of Pediatric Oncology, Children's Cancer Hospital, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mahmoud Hammad
- Department of Pediatric Oncology, Children's Cancer Hospital, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Herbert Pichler
- Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Thomas Lion
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Athanasios Tragiannidis
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Haruko Shima
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Wenbin An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin, China
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, CAMS & PUMC, Tianjin, China
| | - Axel Karow
- Division of Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital, Erlangen, Germany
| | - Roula Farah
- Department of Pediatrics, Lebanese American University Medical Center-Rizk Hospital, Beirut, Lebanon
| | - Maaike Luesink
- Department of Pediatric Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Michael Dworzak
- Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Stephanie Sembill
- Division of Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital, Erlangen, Germany
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Petr Sedlacek
- Department of Pediatric Hematology-Oncology, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Kirk R Schultz
- Division of Hematology/Oncology/BMT, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Krzysztof Kalwak
- Department of Pediatric Hematology, Oncology and BMT, Wroclaw Medical University, Wroclaw, Poland
| | - Birgitta Versluys
- Department of Pediatric Blood and Marrow Transplantation, Princess Máxima Center, Utrecht, The Netherlands
| | - Uma Athale
- Division of Haematology-Oncology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Nobuko Hijiya
- Division of Pediatric Hematology/Oncology/Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Markus Metzler
- Division of Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital, Erlangen, Germany
| | - Meinolf Suttorp
- Department of Pediatric Hematology and Oncology, Medical Faculty, Technical University, Dresden, Germany
| |
Collapse
|
2
|
Ren J, Amoozgar Z, Uccello TP, Lei PJ, Zhao Y, Ho WW, Huang P, Kardian A, Mack SC, Duda DG, Xu L, Jain RK. Targeting EPHB2/ABL1 restores antitumor immunity in preclinical models of ependymoma. Proc Natl Acad Sci U S A 2025; 122:e2319474122. [PMID: 39841145 PMCID: PMC11789170 DOI: 10.1073/pnas.2319474122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
Ependymoma (EPN) is a common form of brain tumor in children, often resistant to available cytotoxic therapies. Molecular profiling studies have led to a better understanding of EPN subtypes and revealed a critical role of oncogenes ZFTA-RELA fusion and EPHB2 in supratentorial ependymoma (ST-EPN). However, the immune system's role in tumor progression and response to therapy remains poorly understood. New treatments for various molecular subtypes of EPN are desperately needed. Using ST-EPN-ZFTA subtype-specific syngeneic mouse models, we found an increased frequency of M2-like tumor-associated macrophages (TAMs), which proportionally increased with tumor size during tumor progression. Transcriptomic profiling of ST-EPN-ZFTA and analysis of a human EPN dataset revealed multiple protein kinases as potential druggable targets. By matching transcriptomic signatures with the target spectrum of FDA-approved drugs, we found that the multikinase inhibitor dasatinib potently inhibited the growth of EPN both in vitro and in vivo, mainly through blocking EPHB2 and ABL1. Treatment with dasatinib reprogrammed the EPN immune microenvironment by polarizing TAMs toward an M1-like phenotype and increasing CD8 T cell activation. Furthermore, dasatinib treatment induced complete regression of established EPN tumors in 78% of the animals and protected survivors against tumor recurrence. Depletion of CD8 cells compromised the durability of EPN responses and reduced overall survival. These data indicate that dasatinib has the potential to be an effective therapy for ST-EPN-ZFTA molecular subgroup of EPN and support further investigation of dasatinib in clinical trials.
Collapse
Affiliation(s)
- Jun Ren
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Zohreh Amoozgar
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Taylor P. Uccello
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Pin-Ji Lei
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Yuhui Zhao
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - William W. Ho
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Peigen Huang
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Alisha Kardian
- Center of Excellence in Neuro-Oncology Sciences, St Jude Children’s Research Hospital, Memphis, TN38105
- Neurobiology and Brain Tumor Program, St Jude Children’s Research Hospital, Memphis, TN38105
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN38105
- Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX77030
| | - Stephen C. Mack
- Center of Excellence in Neuro-Oncology Sciences, St Jude Children’s Research Hospital, Memphis, TN38105
- Neurobiology and Brain Tumor Program, St Jude Children’s Research Hospital, Memphis, TN38105
- Department of Developmental Neurobiology, St Jude Children’s Research Hospital, Memphis, TN38105
- Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX77030
| | - Dan G. Duda
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Lei Xu
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Rakesh K. Jain
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| |
Collapse
|
3
|
Bonada M, Pittarello M, De Fazio E, Gans A, Alimonti P, Slika H, Legnani F, Di Meco F, Tyler B. Pediatric Hemispheric High-Grade Gliomas and H3.3-G34 Mutation: A Review of the Literature on Biological Features and New Therapeutic Strategies. Genes (Basel) 2024; 15:1038. [PMID: 39202398 PMCID: PMC11353413 DOI: 10.3390/genes15081038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Pediatric high-grade glioma (pHGG) encompasses a wide range of gliomas with different genomic, epigenomic, and transcriptomic features. Almost 50% of pHGGs present a mutation in genes coding for histone 3, including the subtype harboring the H3.3-G34 mutation. In this context, histone mutations are frequently associated with mutations in TP53 and ATRX, along with PDGFRA and NOTCH2NL amplifications. Moreover, the H3.3-G34 histone mutation induces epigenetic changes in immune-related genes and exerts modulatory functions on the microenvironment. Also, the functionality of the blood-brain barrier (BBB) has an impact on treatment response. The prognosis remains poor with conventional treatments, thus eliciting the investigation of additional and alternative therapies. Promising molecular targets include PDGFRA amplification, BRAF mutation, EGFR amplification, NF1 loss, and IDH mutation. Considering that pHGGs harboring the H3.3-G34R mutation appear to be more susceptible to immunotherapies (ITs), different options have been recently explored, including immune checkpoint inhibitors, antibody mediated IT, and Car-T cells. This review aims to summarize the knowledge concerning cancer biology and cancer-immune cell interaction in this set of pediatric gliomas, with a focus on possible therapeutic options.
Collapse
Affiliation(s)
- Marta Bonada
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
| | - Matilde Pittarello
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy;
| | - Emerson De Fazio
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy;
| | - Alessandro Gans
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
- ASST Ovest Milanese, Neurology and Stroke Unit, Neuroscience Department, 20025 Legnano, Italy
| | - Paolo Alimonti
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA;
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Federico Legnani
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
| | - Francesco Di Meco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| |
Collapse
|
4
|
Ruiz J, Kelly RK, Aplenc R, Laetsch TW, Seif AE. Absolute neutrophil count clinical trial eligibility criteria for pediatric oncology phase I and phase I/II trials by sponsorship. Pediatr Blood Cancer 2024; 71:e30925. [PMID: 38409529 DOI: 10.1002/pbc.30925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/03/2024] [Accepted: 02/09/2024] [Indexed: 02/28/2024]
Abstract
Normal absolute neutrophil count (ANC) variations, as seen with Duffy-null associated neutrophil count (DANC), are not accounted for in trial eligibility, which may contribute to racial enrollment disparities. We describe ANC eligibility for pediatric oncology phase I/II clinical trials according to primary sponsorship from 2010 to 2023 using ClinicalTrials.gov. Out of 438 trials, 20% were industry-sponsored. Total 17% of trials required ANC ≥1500 cells/μL for enrollment; however, industry-sponsored trials were significantly more likely to require ANC ≥1500 cells/μL than non-industry-sponsored trials (odds ratio 2.53, 95% confidence interval: 1.39-4.62; p < .001). These data suggest laboratory exclusion criteria are one possible mechanism for pediatric clinical trial enrollment disparities.
Collapse
Affiliation(s)
- Jenny Ruiz
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rebecca K Kelly
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Richard Aplenc
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Theodore W Laetsch
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alix E Seif
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Pieters R, Mullighan CG, Hunger SP. Advancing Diagnostics and Therapy to Reach Universal Cure in Childhood ALL. J Clin Oncol 2023; 41:5579-5591. [PMID: 37820294 PMCID: PMC10730082 DOI: 10.1200/jco.23.01286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/24/2023] [Accepted: 08/10/2023] [Indexed: 10/13/2023] Open
Abstract
Systemic combination chemotherapy and intrathecal chemotherapy markedly increased the survival rate of children with ALL. In the past two decades, the use of minimal (measurable) residual disease (MRD) measurements early in therapy improved risk group stratification with subsequent treatment intensifications for patients at high risk of relapse, and enabled a reduction of treatment for low-risk patients. The recent development of more sensitive MRD technologies may further affect risk stratification. Molecular genetic profiling has led to the discovery of many new subtypes and their driver genetic alterations. This increased our understanding of the biological basis of ALL, improved risk classification, and enabled implementation of precision medicine. In the past decade, immunotherapies, including bispecific antibodies, antibody-drug conjugates, and cellular therapies directed against surface proteins, led to more effective and less toxic therapies, replacing intensive chemotherapy courses and allogeneic stem-cell transplantation in patients with relapsed and refractory ALL, and are now being tested in newly diagnosed patients. It has taken 50-60 years to increase the cure rate in childhood ALL from 0% to 90% by stepwise improvements in chemotherapy. This review provides an overview of how the developments over the past 10-15 years mentioned above have significantly changed the diagnostic and treatment approach in ALL, and discusses how the integrated use of molecular and immunotherapeutic insights will very likely direct efforts to cure those children with ALL who are not cured today, and improve the quality of life for survivors who should have decades of life ahead. Future efforts must focus on making effective, yet very expensive, new technologies and therapies available to children with ALL worldwide.
Collapse
Affiliation(s)
- Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Charles G. Mullighan
- Department of Pathology and Hematological Malignancies Program, Comprehensive Cancer Center, St Jude Children's Research Hospital, Memphis, TN
| | - Stephen P. Hunger
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
6
|
Ata F, Benkhadra M, Ghasoub R, Fernyhough LJ, Omar NE, Nashwan AJ, Aldapt MB, Mushtaq K, Kassem NA, Yassin MA. Tyrosine Kinase Inhibitors in pediatric chronic myeloid leukemia: a focused review of clinical trials. Front Oncol 2023; 13:1285346. [PMID: 38188307 PMCID: PMC10769570 DOI: 10.3389/fonc.2023.1285346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
Tyrosine Kinase Inhibitors (TKIs) is revolutionizing the management of pediatric Chronic Myeloid Leukemia (CML), offering alternatives to Allogeneic Hematopoietic Stem Cell Transplantation (AHSCT). We conducted a comprehensive review of 16 Randomized Controlled Trials (RCTs) encompassing 887 pediatric CML patients treated with TKIs including Imatinib, Dasatinib, and Nilotinib. The median patient age ranged from 6.5 to 14 years, with a median white blood cell count of 234 x 10^9/uL, median hemoglobin level of 9.05 g/dL, and median platelet count of 431.5 x 10^9/µL. Imatinib seems to be predominant first line TKI, with the most extensive safety and efficacy data. BCR::ABL response rates below 10% ranged from 60% to 78%, CCyR at 24 months ranged from 62% to 94%, and PFS showed variability from 56.8% to 100%, albeit with differing analysis timepoints. The Safety profile of TKIs was consistent with the known safety profile in adults. With the availability of three TKIs as first line options, multiple factors should be considered when selecting first line TKI, including drug formulation, administration, comorbidities, and financial issues. Careful monitoring of adverse events, especially in growing children, should be considered in long term follow-up clinical trials.
Collapse
Affiliation(s)
- Fateen Ata
- Department of Endocrinology and Metabolism, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Maria Benkhadra
- Pharmacy Department, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Rola Ghasoub
- Pharmacy Department, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Liam J. Fernyhough
- Department of Medical Education, Weill Cornell Medicine Qatar, Doha, Qatar
| | - Nabil E. Omar
- Pharmacy Department, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- Health Sciences Program, Clinical and Population Health Research, College of Pharmacy, Qatar University, Doha, Qatar
| | | | - Mahmood B. Aldapt
- Department of Medicine, Unity Hospital/Rochester Regional Health, Rochester, NY, United States
| | - Kamran Mushtaq
- Department of Gastroenterology, University Hospital Southampton, Southampton, United Kingdom
| | - Nancy A. Kassem
- Pharmacy Department, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mohamed A. Yassin
- Department of Medical Oncology/Hematology, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
7
|
Gastberger K, Fincke VE, Mucha M, Siebert R, Hasselblatt M, Frühwald MC. Current Molecular and Clinical Landscape of ATRT - The Link to Future Therapies. Cancer Manag Res 2023; 15:1369-1393. [PMID: 38089834 PMCID: PMC10712249 DOI: 10.2147/cmar.s379451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/28/2023] [Indexed: 10/16/2024] Open
Abstract
ATRT is a highly aggressive and rare pediatric CNS tumor of very young children. Its genetic hallmark is bi-allelic inactivation of SMARCB1 encoding INI1. Rarely SMARCA4 encoding BRG1 is affected. Up to 30% are associated with constitutional heterozygous pathogenic variants in one of the two genes, giving rise to the Rhabdoid-Tumor-Predisposition-Syndromes (RTPS) 1 and 2. Characteristic DNA methylation profiles distinguish ATRT from other SMARCB1-deficient entities. Three distinct subtypes ATRT-MYC, -TYR, and -SHH are on record. ATRT-SHH may be further divided into the subgroups ATRT-SHH1A, -SHH1B, and -SHH2. The cure of ATRT remains challenging, notwithstanding an increasing understanding of molecular pathomechanisms and genetic background. The implementation of multimodal institutional treatment protocols has improved prognosis. Regardless of treatment approaches, clinical risk factors such as age, metastases, and DNA methylation subtype affect survival probability. We provide a critical appraisal of current conventional multimodal regimens and emerging targeted treatment approaches investigated in clinical trials and entity-specific registries. Intense treatment approaches featuring radiotherapy (RT) and high-dose chemotherapy (HDCT) face the difficulty of balancing tumor control and treatment-related toxicity. Current approaches focus on minimizing radiation fields by proton beam therapy or to withhold RT in HDCT-only approaches. Still, a 40-75% relapse rate upon first-line treatment reveals the need for novel treatment strategies in primary and even more in recurrent/refractory (r/r) disease. Among targeted treatments, immune checkpoint inhibitors and epigenetically active agents appear most promising. Success remains limited in single agent approaches. We hypothesize that mechanism-informed combination therapy will enhance response, as the low mutational burden of ATRT may contribute to acquiring resistance to single targeted agents. As DNA methylation group-specific gene expression profiles appear to influence response to distinct agents, the future treatment of ATRT should respect clinical and biological heterogeneity in risk group adjusted treatment protocols.
Collapse
Affiliation(s)
- Katharina Gastberger
- Pediatrics and Adolescent Medicine, Swabian Children’s Cancer Center, University Medical Center Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Victoria E Fincke
- Pediatrics and Adolescent Medicine, Swabian Children’s Cancer Center, University Medical Center Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Marlena Mucha
- Pediatrics and Adolescent Medicine, Swabian Children’s Cancer Center, University Medical Center Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Michael C Frühwald
- Pediatrics and Adolescent Medicine, Swabian Children’s Cancer Center, University Medical Center Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| |
Collapse
|
8
|
Akshintala S, Sundby RT, Bernstein D, Glod JW, Kaplan RN, Yohe ME, Gross AM, Derdak J, Lei H, Pan A, Dombi E, Palacio-Yance I, Herrera KR, Miettinen MM, Chen HX, Steinberg SM, Helman LJ, Mascarenhas L, Widemann BC, Navid F, Shern JF, Heske CM. Phase I trial of Ganitumab plus Dasatinib to Cotarget the Insulin-Like Growth Factor 1 Receptor and Src Family Kinase YES in Rhabdomyosarcoma. Clin Cancer Res 2023; 29:3329-3339. [PMID: 37398992 PMCID: PMC10529967 DOI: 10.1158/1078-0432.ccr-23-0709] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/05/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE Antibodies against insulin-like growth factor (IGF) type 1 receptor have shown meaningful but transient tumor responses in patients with rhabdomyosarcoma (RMS). The SRC family member YES has been shown to mediate IGF type 1 receptor (IGF-1R) antibody acquired resistance, and cotargeting IGF-1R and YES resulted in sustained responses in murine RMS models. We conducted a phase I trial of the anti-IGF-1R antibody ganitumab combined with dasatinib, a multi-kinase inhibitor targeting YES, in patients with RMS (NCT03041701). PATIENTS AND METHODS Patients with relapsed/refractory alveolar or embryonal RMS and measurable disease were eligible. All patients received ganitumab 18 mg/kg intravenously every 2 weeks. Dasatinib dose was 60 mg/m2/dose (max 100 mg) oral once daily [dose level (DL)1] or 60 mg/m2/dose (max 70 mg) twice daily (DL2). A 3+3 dose escalation design was used, and maximum tolerated dose (MTD) was determined on the basis of cycle 1 dose-limiting toxicities (DLT). RESULTS Thirteen eligible patients, median age 18 years (range 8-29) enrolled. Median number of prior systemic therapies was 3; all had received prior radiation. Of 11 toxicity-evaluable patients, 1/6 had a DLT at DL1 (diarrhea) and 2/5 had a DLT at DL2 (pneumonitis, hematuria) confirming DL1 as MTD. Of nine response-evaluable patients, one had a confirmed partial response for four cycles, and one had stable disease for six cycles. Genomic studies from cell-free DNA correlated with disease response. CONCLUSIONS The combination of dasatinib 60 mg/m2/dose daily and ganitumab 18 mg/kg every 2 weeks was safe and tolerable. This combination had a disease control rate of 22% at 5 months.
Collapse
Affiliation(s)
- Srivandana Akshintala
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - R. Taylor Sundby
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Donna Bernstein
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - John W. Glod
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Rosandra N. Kaplan
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Marielle E. Yohe
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, Maryland
| | - Andrea M. Gross
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Joanne Derdak
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Alexander Pan
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Isabel Palacio-Yance
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Kailey R. Herrera
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Markku M. Miettinen
- Laboratory of Pathology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Helen X. Chen
- Cancer Therapy Evaluation Program (CTEP), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Seth M. Steinberg
- Biostatistics and Data Management, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Lee J. Helman
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles (CHLA), Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- The Osteosarcoma Institute, Dallas, Texas
| | - Leo Mascarenhas
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles (CHLA), Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brigitte C. Widemann
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Fariba Navid
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles (CHLA), Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| | - Christine M. Heske
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland
| |
Collapse
|
9
|
Yoshimura S, Panetta JC, Hu J, Li L, Gocho Y, Du G, Umezawa A, Karol SE, Pui CH, Mullighan CG, Konopleva M, Stock W, Teachey DT, Jain N, Yang JJ. Preclinical pharmacokinetic and pharmacodynamic evaluation of dasatinib and ponatinib for the treatment of T-cell acute lymphoblastic leukemia. Leukemia 2023; 37:1194-1203. [PMID: 37076694 PMCID: PMC10347458 DOI: 10.1038/s41375-023-01900-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023]
Abstract
LCK is a novel therapeutic target in ~40% of T-cell acute lymphoblastic leukemia (T-ALL), and dasatinib and ponatinib can act as LCK inhibitors with therapeutic effects. We herein report a comprehensive preclinical pharmacokinetic and pharmacodynamic evaluation of dasatinib and ponatinib in LCK-activated T-ALL. In 51 human T-ALL cases, these two drugs showed similar patterns of cytotoxic activity, with ponatinib being slightly more potent. Given orally in mice, ponatinib was associated with slower clearance with a longer Tmax and higher AUC0-24 h, although maximum pLCK inhibition was comparable between the two drugs. After establishing the exposure-to-response models, we simulated the steady-state pLCK inhibitory effects of each drug at currently approved dosages in humans: dasatinib at 140 mg and ponatinib at 45 mg once daily are both sufficient to achieve >50% pLCK inhibition for 13.0 and 13.9 h/day, respectively, comparable to pharmacodynamic profiles of these agents in BCR::ABL1 leukemias. Moreover, we developed a dasatinib-resistant T-ALL cell line model with LCK T316I mutation, in which ponatinib retained partial activity against LCK. In conclusion, we described the pharmacokinetic and pharmacodynamic profiles of dasatinib and ponatinib as LCK inhibitors in T-ALL, providing critical data for the development of human trials of these agents.
Collapse
Affiliation(s)
- Satoshi Yoshimura
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo, Japan
| | - John C Panetta
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jianzhong Hu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
- Amgen, 1 Amgen Center Drive, Thousand Oaks, CA, USA
| | - Lie Li
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yoshihiro Gocho
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Guoqing Du
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Akihiro Umezawa
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo, Japan
| | - Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marina Konopleva
- Department of Oncology and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wendy Stock
- Department of Medicine Section of Hematology-Oncology, University of Chicago, Chicago, IL, USA
| | - David T Teachey
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
| | - Nitin Jain
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun J Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
10
|
Zou H, Poore B, Brown EE, Qian J, Xie B, Asimakidou E, Razskazovskiy V, Ayrapetian D, Sharma V, Xia S, Liu F, Chen A, Guan Y, Li Z, Wanggou S, Saulnier O, Ly M, Fellows-Mayle W, Xi G, Tomita T, Resnick AC, Mack SC, Raabe EH, Eberhart CG, Sun D, Stronach BE, Agnihotri S, Kohanbash G, Lu S, Herrup K, Rich JN, Gittes GK, Broniscer A, Hu Z, Li X, Pollack IF, Friedlander RM, Hainer SJ, Taylor MD, Hu B. A neurodevelopmental epigenetic programme mediated by SMARCD3-DAB1-Reelin signalling is hijacked to promote medulloblastoma metastasis. Nat Cell Biol 2023; 25:493-507. [PMID: 36849558 PMCID: PMC10014585 DOI: 10.1038/s41556-023-01093-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/17/2023] [Indexed: 03/01/2023]
Abstract
How abnormal neurodevelopment relates to the tumour aggressiveness of medulloblastoma (MB), the most common type of embryonal tumour, remains elusive. Here we uncover a neurodevelopmental epigenomic programme that is hijacked to induce MB metastatic dissemination. Unsupervised analyses of integrated publicly available datasets with our newly generated data reveal that SMARCD3 (also known as BAF60C) regulates Disabled 1 (DAB1)-mediated Reelin signalling in Purkinje cell migration and MB metastasis by orchestrating cis-regulatory elements at the DAB1 locus. We further identify that a core set of transcription factors, enhancer of zeste homologue 2 (EZH2) and nuclear factor I X (NFIX), coordinates with the cis-regulatory elements at the SMARCD3 locus to form a chromatin hub to control SMARCD3 expression in the developing cerebellum and in metastatic MB. Increased SMARCD3 expression activates Reelin-DAB1-mediated Src kinase signalling, which results in a MB response to Src inhibition. These data deepen our understanding of how neurodevelopmental programming influences disease progression and provide a potential therapeutic option for patients with MB.
Collapse
Affiliation(s)
- Han Zou
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Changsha, China
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley Poore
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Emily E Brown
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jieqi Qian
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Evridiki Asimakidou
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Vladislav Razskazovskiy
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Deanna Ayrapetian
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Vaibhav Sharma
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Shunjin Xia
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Fei Liu
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Apeng Chen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Yongchang Guan
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Zhengwei Li
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Olivier Saulnier
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michelle Ly
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Wendy Fellows-Mayle
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guifa Xi
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tadanori Tomita
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephen C Mack
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Eric H Raabe
- Division of Pediatric Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beth E Stronach
- Office of Research, University of Pittsburgh Health Sciences, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - George K Gittes
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alberto Broniscer
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhongliang Hu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Changsha, China
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Robert M Friedlander
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah J Hainer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- John G. Rangos Sr Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Narayan HK, Sheline K, Wong V, Kuo D, Choo S, Yoon J, Leger K, Kutty S, Fradley M, Tremoulet A, Ky B, Armenian S, Guha A. Cardiovascular toxicities with pediatric tyrosine kinase inhibitor therapy: An analysis of adverse events reported to the Food and Drug Administration. Pediatr Blood Cancer 2023; 70:e30059. [PMID: 36385736 DOI: 10.1002/pbc.30059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022]
Abstract
We sought to examine cardiovascular toxicities associated with tyrosine kinase inhibitors in pediatrics. We examined 1624 pediatric adverse events with imatinib, dasatinib, sorafenib, pazopanib, crizotinib, and ruxolitinib reported to the Food and Drug Administration between January 1, 2015, and August 14, 2020. There were 102 cardiovascular event reports. Hypertension was the most commonly reported cardiovascular event and was most frequently associated with sorafenib and pazopanib. The presence of infection increased the reporting odds of cardiovascular events overall and specifically cardiac arrest, heart failure, and hypertension. These data provide early insight into cardiovascular toxicities with tyrosine kinase inhibitor use in pediatrics.
Collapse
Affiliation(s)
- Hari K Narayan
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Karyn Sheline
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Victor Wong
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Dennis Kuo
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Sun Choo
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Janet Yoon
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Kasey Leger
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Shelby Kutty
- Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael Fradley
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adriana Tremoulet
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Bonnie Ky
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saro Armenian
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Avirup Guha
- Cardio-Oncology Program, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
12
|
Bernsen EC, Hogenes VJ, Nuijen B, Hanff LM, Huitema ADR, Diekstra MHM. Practical Recommendations for the Manipulation of Kinase Inhibitor Formulations to Age-Appropriate Dosage Forms. Pharmaceutics 2022; 14:2834. [PMID: 36559327 PMCID: PMC9782008 DOI: 10.3390/pharmaceutics14122834] [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: 10/28/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Over 75 kinase inhibitors (KIs) have been approved for the treatment of various cancers. KIs are orally administrated but mostly lack pediatric age-appropriate dosage forms or instructions for dose manipulation. This is highly problematic for clinical practice in pediatric oncology, as flexible oral formulations are essential to individually set dosages and to adjust it to a child's swallowability. Most KIs are poorly soluble, categorized in Biopharmaceutics Classification System (BCS) class II or IV, and improperly manipulating the KI formulation can alter pharmacokinetics and jeopardize KI drug safety and efficacy. Therefore, the goals of this review were to provide practical recommendations for manipulating the formulation of the 15 most frequently used KIs in pediatric oncology (i.e., bosutinib, cabozantinib, cobimetinib, crizotinib, dabrafenib, dasatinib, entrectinib, imatinib, larotrectinib, nilotinib, ponatinib, ruxolitinib, selumetinib, sunitinib and trametinib) based on available literature studies and fundamental drug characteristics and to establish a decision tool that supports decisions regarding formulation manipulation of solid oral dosages of KIs that have been or will be licensed (for adult and/or pediatric cancers) but are not included in this review.
Collapse
Affiliation(s)
- Emma C. Bernsen
- Princess Máxima Center for Pediatric Oncology, Department of Pharmacology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Valery J. Hogenes
- Princess Máxima Center for Pediatric Oncology, Department of Pharmacology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Lidwien M. Hanff
- Princess Máxima Center for Pediatric Oncology, Department of Pharmacology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Alwin D. R. Huitema
- Princess Máxima Center for Pediatric Oncology, Department of Pharmacology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Meta H. M. Diekstra
- Princess Máxima Center for Pediatric Oncology, Department of Pharmacology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
13
|
Yu L, Zhang J, Li Y. Effects of microenvironment in osteosarcoma on chemoresistance and the promise of immunotherapy as an osteosarcoma therapeutic modality. Front Immunol 2022; 13:871076. [PMID: 36311748 PMCID: PMC9608329 DOI: 10.3389/fimmu.2022.871076] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/28/2022] [Indexed: 12/02/2022] Open
Abstract
Osteosarcoma (OS) is one of the most common primary malignant tumors originating in bones. Its high malignancy typically manifests in lung metastasis leading to high mortality. Although remarkable advances in surgical resection and neoadjuvant chemotherapy have lengthened life expectancy and greatly improved the survival rate among OS patients, no further breakthroughs have been achieved. It is challenging to treat patients with chemoresistant tumors and distant metastases. Recent studies have identified a compelling set of links between hypoxia and chemotherapy failure. Here, we review the evidence supporting the positive effects of hypoxia in the tumor microenvironment (TME). In addition, certain anticancer effects of immune checkpoint inhibitors have been demonstrated in OS preclinical models. Continued long-term observation in clinical trials is required. In the present review, we discuss the mutualistic effects of the TME in OS treatment and summarize the mechanisms of immunotherapy and their interaction with TME when used to treat OS. We also suggest that immunotherapy, a new comprehensive and potential antitumor approach that stimulates an immune response to eliminate tumor cells, may represent an innovative approach for the development of a novel treatment regimen for OS patients.
Collapse
|
14
|
Origin and Therapies of Osteosarcoma. Cancers (Basel) 2022; 14:cancers14143503. [PMID: 35884563 PMCID: PMC9322921 DOI: 10.3390/cancers14143503] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Osteosarcoma is the most common malignant bone tumor in children, with a 5-year survival rate ranging from 70% to 20% depending on the aggressiveness of the disease. The current treatments have not evolved over the past four decades due in part to the genetic complexity of the disease and its heterogeneity. This review will summarize the current knowledge of OS origin, diagnosis and therapies. Abstract Osteosarcoma (OS) is the most frequent primary bone tumor, mainly affecting children and young adults. Despite therapeutic advances, the 5-year survival rate is 70% but drastically decreases to 20–30% for poor responders to therapies or for patients with metastasis. No real evolution of the survival rates has been observed for four decades, explained by poor knowledge of the origin, difficulties related to diagnosis and the lack of targeted therapies for this pediatric tumor. This review will describe a non-exhaustive overview of osteosarcoma disease from a clinical and biological point of view, describing the origin, diagnosis and therapies.
Collapse
|
15
|
Finch ER, Janke LJ, Li L, Payton MA, Jenkins DA, Crews KR, Relling MV, Karol SE. Dasatinib does not exacerbate dexamethasone-induced osteonecrosis in murine models of acute lymphoblastic leukemia therapy. Pediatr Blood Cancer 2022; 69:e29490. [PMID: 34866312 PMCID: PMC8860849 DOI: 10.1002/pbc.29490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/26/2021] [Accepted: 11/10/2021] [Indexed: 01/22/2023]
Abstract
INTRODUCTION There are clinical reports that the incorporation of dasatinib may increase the frequency of osteonecrosis in acute lymphoblastic leukemia (ALL) treatment regimens. No rigorous testing of this hypothesis is available to guide clinicians. METHODS We tested whether oral dasatinib increased the frequency of dexamethasone-induced osteonecrosis in a murine model and tested its effects on dexamethasone's antileukemic efficacy in a murine BCR-ABL+ model of ALL. RESULTS Dasatinib did not change the frequency of osteonecrosis (p = .99) nor of arteriopathy (p = .36) in dexamethasone-treated mice when given at dosages that achieved clinically relevant steady-state dasatinib plasma concentrations of 53.1 ng/ml (95% CI: 43.5-57.3 ng/ml). These dasatinib exposures were not associated with increased dexamethasone plasma exposure in nonleukemia-bearing mice. These same dosages were not associated with any decrement in antileukemic efficacy of dexamethasone in a responsive BCR-ABL+ model of ALL. CONCLUSIONS Based on the results of our preclinical murine studies, we conclude that dasatinib is unlikely to increase the osteonecrotic effects of dexamethasone in ALL regimens.
Collapse
Affiliation(s)
- Emily R. Finch
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Laura J. Janke
- Department of Pathology, Division of Comparative Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Lie Li
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Monique A. Payton
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - David A. Jenkins
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kristine R. Crews
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Seth E. Karol
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
16
|
Kraveka JM, Lewis EC, Bergendahl G, Ferguson W, Oesterheld J, Kim E, Nagulapally AB, Dykema KJ, Brown VI, Roberts WD, Mitchell D, Eslin D, Hanson D, Isakoff MS, Wada RK, Harrod VL, Rawwas J, Hanna G, Hendricks WPD, Byron SA, Snuderl M, Serrano J, Trent JM, Saulnier Sholler GL. A pilot study of genomic-guided induction therapy followed by immunotherapy with difluoromethylornithine maintenance for high-risk neuroblastoma. Cancer Rep (Hoboken) 2022; 5:e1616. [PMID: 35355452 PMCID: PMC9675391 DOI: 10.1002/cnr2.1616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/16/2022] [Accepted: 02/27/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Survival for patients with high-risk neuroblastoma (HRNB) remains poor despite aggressive multimodal therapies. AIMS To study the feasibility and safety of incorporating a genomic-based targeted agent to induction therapy for HRNB as well as the feasibility and safety of adding difluoromethylornithine (DFMO) to anti-GD2 immunotherapy. METHODS Twenty newly diagnosed HRNB patients were treated on this multicenter pilot trial. Molecular tumor boards selected one of six targeted agents based on tumor-normal whole exome sequencing and tumor RNA-sequencing results. Treatment followed standard upfront HRNB chemotherapy with the addition of the selected targeted agent to cycles 3-6 of induction. Following consolidation, DFMO (750 mg/m2 twice daily) was added to maintenance with dinutuximab and isotretinoin, followed by continuation of DFMO alone for 2 years. DNA methylation analysis was performed retrospectively and compared to RNA expression. RESULTS Of the 20 subjects enrolled, 19 started targeted therapy during cycle 3 and 1 started during cycle 5. Eighty-five percent of subjects met feasibility criteria (receiving 75% of targeted agent doses). Addition of targeted agents did not result in toxicities requiring dose reduction of chemotherapy or permanent discontinuation of targeted agent. Following standard consolidation, 15 subjects continued onto immunotherapy with DFMO. This combination was well-tolerated and resulted in no unexpected adverse events related to DFMO. CONCLUSION This study demonstrates the safety and feasibility of adding targeted agents to standard induction therapy and adding DFMO to immunotherapy for HRNB. This treatment regimen has been expanded to a Phase II trial to evaluate efficacy.
Collapse
Affiliation(s)
| | - Elizabeth C. Lewis
- Wayne State University School of MedicineDetroitMichiganUSA,Levine Children's Hospital, Atrium HealthCharlotteNorth CarolinaUSA
| | | | | | | | - Elizabeth Kim
- Levine Children's Hospital, Atrium HealthCharlotteNorth CarolinaUSA,Wesleyan UniversityMiddletownConnecticutUSA
| | | | - Karl J. Dykema
- Levine Children's Hospital, Atrium HealthCharlotteNorth CarolinaUSA
| | - Valerie I. Brown
- Penn State Children's Hospital at the Milton S. Hershey Medical Center and Penn State College of MedicineHersheyPennsylvaniaUSA
| | - William D. Roberts
- Rady Children's Hospital San Diego and UC San Diego School of MedicineSan DiegoCaliforniaUSA
| | - Deanna Mitchell
- Helen DeVos Children's Hospital at Spectrum HealthGrand RapidsMichiganUSA
| | - Don Eslin
- St. Joseph's Children's HospitalTampaFloridaUSA
| | - Derek Hanson
- Hackensack University Medical CenterHackensackNew JerseyUSA
| | - Michael S. Isakoff
- Center for Cancer and Blood DisordersConnecticut Children's Medical CenterHartfordConnecticutUSA
| | - Randal K. Wada
- Kapiolani Medical Center for Women & ChildrenHonoluluHawaiiUSA
| | | | - Jawhar Rawwas
- Children's Hospitals and Clinics of MinnesotaMinneapolisMinnesotaUSA
| | - Gina Hanna
- Orlando Health Cancer InstituteOrlandoFloridaUSA
| | | | - Sara A. Byron
- Translational Genomics Research Institute (TGen)PhoenixArizonaUSA
| | - Matija Snuderl
- NYU Langone Health and NYU Grossman School of MedicineNew York CityNew YorkUSA
| | - Jonathan Serrano
- NYU Langone Health and NYU Grossman School of MedicineNew York CityNew YorkUSA
| | - Jeffrey M. Trent
- Translational Genomics Research Institute (TGen)PhoenixArizonaUSA
| | | |
Collapse
|
17
|
Elsayed HRH, El-Gamal R, Rabei MR, Elhadidy MG, Hamed S, Othman BH, Elshaer MMA, Sedky MK, Hassan ATAE, El-Nablaway M. Enhanced Autophagic Flux, Suppressed Apoptosis and Reduced Macrophage Infiltration by Dasatinib in Kidneys of Obese Mice. Cells 2022; 11:cells11040746. [PMID: 35203394 PMCID: PMC8869974 DOI: 10.3390/cells11040746] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 01/15/2023] Open
Abstract
Obesity causes renal changes (ORC), characterized by defective renal autophagy, lipogenesis, enhanced macrophage infiltration and apoptosis. We hypothesize that Dasatinib, a tyrosine kinase inhibitor, may ameliorate changes associated with obesity. We the mice with either Obesogenic diet (OD) or a standard basal diet. After 12 weeks, the mice received either vehicle or Dasatinib 4 mg/kg/d for an additional four weeks. We examined serum creatinine, urea, lipid profile and renal cortical mRNA expression for lipogenesis marker SREBP1, inflammatory macrophage marker iNOS and fibrosis markers; TGFβ and PDGFA genes; immunohistochemical (IHC) staining for CD68; inflammatory macrophage marker and ASMA; fibrosis marker, LC3 and SQSTM1/P62; autophagy markers and western blotting (WB) for caspase-3; and, as an apoptosis marker, LC3II/I and SQSTM1/P62 in addition to staining for H&E, PAS, Sirius red and histopathological scoring. Dasatinib attenuated renal cortical mRNA expression for SREBP1, iNOS, PDGFA and TGFβ and IHC staining for CD68, ASMA and SQSTM1/P62 and WB for caspase-3 and SQSTM1/P62, while elevating LC3 expression. Moreover, Dasatinib ameliorated ORC; glomerulosclerosis, glomerular expansion, tubular dilatation, vacuolation and casts; inflammatory cellular infiltration; and fibrosis. Dasatinib is a promising therapy for ORC by correcting autophagy impairment, attenuating lipogenesis, apoptosis and macrophage infiltration by inducing antifibrotic activity.
Collapse
Affiliation(s)
- Hassan Reda Hassan Elsayed
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Anatomy, Faculty of Medicine, Horus University, New Damietta 34517, Egypt
- Correspondence: ; Tel.: +20-122-9310-701
| | - Randa El-Gamal
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (R.E.-G.); (M.E.-N.)
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohammed R. Rabei
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (M.R.R.); (M.G.E.)
- Department of Physiology, Faculty of Medicine, King Salman International University, South Sinai 46511, Egypt
| | - Mona G. Elhadidy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (M.R.R.); (M.G.E.)
- Department of Medical Physiology, College of Medicine, Al-Baha University, Al-Baha 61008, Saudi Arabia
| | - Shereen Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Basma H. Othman
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed Mahmoud Abdelraheem Elshaer
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
- Department of Clinical Pharmacology, Faculty of Medicine, King Salman International University, South Sinai 46511, Egypt
| | - Mostafa Khaled Sedky
- Department of Surgery, Faculty of Medicine, King Salman International University, South Sinai 46511, Egypt; (M.K.S.); (A.T.A.E.H.)
| | - Ahmed Tarek Abd Elbaset Hassan
- Department of Surgery, Faculty of Medicine, King Salman International University, South Sinai 46511, Egypt; (M.K.S.); (A.T.A.E.H.)
| | - Mohammad El-Nablaway
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (R.E.-G.); (M.E.-N.)
- Department of Medical Biochemistry, College of Medicine, Almaarefa University, Riyadh 71666, Saudi Arabia
| |
Collapse
|
18
|
Liu LY, Teng JMC, Spunt SL, Strelo JL, Kwong BY, Zaba LC. Dermatologic toxicities of targeted antineoplastic agents and immune checkpoint inhibitor therapy in pediatric patients: A systematic review. Pediatr Blood Cancer 2021; 68:e29346. [PMID: 34569142 DOI: 10.1002/pbc.29346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 11/09/2022]
Abstract
Cutaneous adverse events (cAEs) from targeted antineoplastic agents and immune checkpoint inhibitors are common in children with cancer and may lead to dose reduction or cessation of critical oncologic treatment. Timely diagnosis and proper management of cAEs in pediatric oncology patients is essential to optimize ongoing cancer-directed therapy and improve quality of life. This systematic review of published studies summarizes dermatologic toxicities to targeted anticancer treatments and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Lucy Y Liu
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Joyce M C Teng
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA.,Department of Dermatology, Division of Pediatric Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Sheri L Spunt
- Department of Pediatrics, Division of Hematology/Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Jenna L Strelo
- Cutaneous Oncology, Stanford University Medical Center and Cancer Institute, Stanford, California, USA
| | - Bernice Y Kwong
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA.,Cutaneous Oncology, Stanford University Medical Center and Cancer Institute, Stanford, California, USA
| | - Lisa C Zaba
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA.,Cutaneous Oncology, Stanford University Medical Center and Cancer Institute, Stanford, California, USA
| |
Collapse
|
19
|
Abstract
Dasatinib is an oral, once-daily tyrosine kinase inhibitor used in the treatment of chronic myeloid leukaemia and Philadelphia chromosome-positive acute lymphoblastic leukaemia. Dasatinib is rapidly absorbed, with the time for maximal serum concentration varying between 0.25 and 1.5 h. Oral absorption is not affected by food. The absolute bioavailability of dasatinib in humans is unknown due to the lack of an intravenous formulation preventing calculation of the reference exposure. Dasatinib is eliminated through cytochrome P450 (CYP) 3A4-mediated metabolism, with a terminal half-life of 3-4 h. Based on total radioactivity, only 20% of the oral dose (100 mg) is recovered unchanged in faeces (19%, including potential non-absorption) and urine (1%) after 168 h. Dasatinib pharmacokinetics are not influenced by age (children, and adults up to 86 years of age), race and renal insufficiency. Dasatinib absorption is decreased by pH-modifying agents (antacids, H2-receptor blockers, proton pump inhibitors), and dasatinib is also subject to drug interactions with CYP3A4 inducers or inhibitors.
Collapse
|
20
|
Yang F, Zhang L, Zhao BB, Zhang JL, Liu XT, Li X, Tang BH, Zhou Y, Yang XM, van den Anker J, Zhu XF, Zhao W. Population Pharmacokinetics and Safety of Dasatinib in Chinese Children with Core-Binding Factor Acute Myeloid Leukemia. Clin Pharmacokinet 2021; 61:71-81. [PMID: 34240339 DOI: 10.1007/s40262-021-01054-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Dasatinib, an orally administered Src-family kinase inhibitor, is combined with the standard chemotherapeutic regimen to enhance antineoplastic activity against core-binding factor acute myeloid leukemia (CBF-AML) in adults; however, limited data are available for use in children. In the present study, we studied the pharmacokinetics and safety of dasatinib in children. METHODS Dasatinib (60 or 80 mg/m2 once daily) was administered to 20 children with CBF-AML. Blood samples were collected and drug concentrations were quantified by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Population pharmacokinetic analysis and Monte-Carlo simulations were performed using NONMEM software, and safety analyses were assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.0 (NCT03844360). RESULTS Twenty pediatric patients (3.3-14.4 years of age) were included, and a total of 40 dasatinib concentrations were available for population pharmacokinetic analysis. The mean (standard deviation) of the estimated area under the concentration-time curve extrapolated to steady state (AUCss) of dasatinib 60 and 80 mg/m2 was 366.1 (146.6) ng·h/mL and 425.3 (150.7) ng·h/mL, respectively. The majority of adverse events were grade 1/2 in severity, including thrombocytopenia, rash, and pain in the extremities. The estimated cumulative incidence of complete remission and complete molecular response were 95.0% and 75.5%, respectively. CONCLUSIONS The population pharmacokinetics of orally administered dasatinib were evaluated in pediatric CBF-AML patients. The AUCss of dasatinib (80 mg/m2) in CBF-AML pediatric patients was similar to those of dasatinib (100 mg) in adult patients. Dasatinib is well-tolerated in pediatric patients with CBF-AML.
Collapse
Affiliation(s)
- Fan Yang
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Zhang
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Bei-Bei Zhao
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Jing-Liao Zhang
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Xi-Ting Liu
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue Li
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin-Mei Yang
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA.,Departments of Pediatrics, Pharmacology and Physiology, Genomics and Precision Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA.,Department of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Xiao-Fan Zhu
- Department of Pediatrics, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China. .,Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China. .,Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, No.44, Wenhua West Road, Jinan, Shandong, China.
| |
Collapse
|
21
|
Phillips LN, Hijiya N. Tyrosine Kinase Inhibitors and Beyond for Chronic Myeloid Leukemia in Children. Paediatr Drugs 2021; 23:241-251. [PMID: 33899163 DOI: 10.1007/s40272-021-00446-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 12/20/2022]
Abstract
Chronic myeloid leukemia (CML) is rare in children but presents a unique challenge as recent drug innovations have turned CML into a chronic disease with implications for treatment into adulthood. With the approval of newer-generation tyrosine kinase inhibitors (TKIs) in addition to imatinib, providers have more options for the treatment of chronic-phase CML (CML-CP) in children. The second-generation TKIs approved for use in children, nilotinib and dasatinib, have higher response rates than first-generation imatinib; however, overall survival rates appear to be the same. Even more options may soon become available with ongoing investigations into the use of bosutinib and ponatinib and other new agents in children. Possible long-term side effects of TKIs, including growth failure, should be carefully acknowledged by the treating provider. Although these known associations may not preclude treatment, providers should be aware of them to guide their management of pediatric patients with CML being treated long term with TKI therapy. Treatment-free remission is a desired goal for pediatric patients and providers alike, but current recommendations are for attempts at achieving this to be restricted to clinical study settings.
Collapse
Affiliation(s)
- Lia N Phillips
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Nobuko Hijiya
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Gocho Y, Liu J, Hu J, Yang W, Dharia NV, Zhang J, Shi H, Du G, John A, Lin TN, Hunt J, Huang X, Ju B, Rowland L, Shi L, Maxwell D, Smart B, Crews KR, Yang W, Hagiwara K, Zhang Y, Roberts K, Wang H, Jabbour E, Stock W, Eisfelder B, Paietta E, Newman S, Roti G, Litzow M, Easton J, Zhang J, Peng J, Chi H, Pounds S, Relling MV, Inaba H, Zhu X, Kornblau S, Pui CH, Konopleva M, Teachey D, Mullighan CG, Stegmaier K, Evans WE, Yu J, Yang JJ. Network-based systems pharmacology reveals heterogeneity in LCK and BCL2 signaling and therapeutic sensitivity of T-cell acute lymphoblastic leukemia. NATURE CANCER 2021; 2:284-299. [PMID: 34151288 PMCID: PMC8208590 DOI: 10.1038/s43018-020-00167-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy, and novel therapeutics are much needed. Profiling patient leukemia' drug sensitivities ex vivo, we discovered that 44.4% of childhood and 16.7% of adult T-ALL cases exquisitely respond to dasatinib. Applying network-based systems pharmacology analyses to examine signal circuitry, we identified preTCR-LCK activation as the driver of dasatinib sensitivity, and T-ALL-specific LCK dependency was confirmed in genome-wide CRISPR-Cas9 screens. Dasatinib-sensitive T-ALLs exhibited high BCL-XL and low BCL2 activity and venetoclax resistance. Discordant sensitivity of T-ALL to dasatinib and venetoclax is strongly correlated with T-cell differentiation, particularly with the dynamic shift in LCK vs. BCL2 activation. Finally, single-cell analysis identified leukemia heterogeneity in LCK and BCL2 signaling and T-cell maturation stage, consistent with dasatinib response. In conclusion, our results indicate that developmental arrest in T-ALL drives differential activation of preTCR-LCK and BCL2 signaling in this leukemia, providing unique opportunities for targeted therapy.
Collapse
Affiliation(s)
- Yoshihiro Gocho
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jingjing Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jianzhong Hu
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wentao Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jingliao Zhang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hao Shi
- Department of Immunology,, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Guoqing Du
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - August John
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ting-Nien Lin
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeremy Hunt
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xin Huang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lauren Rowland
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lei Shi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dylan Maxwell
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandon Smart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kristine R Crews
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kohei Hagiwara
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Kathryn Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hong Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy Stock
- University of Chicago Medical Center, Chicago, IL, USA
| | | | | | - Scott Newman
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Giovanni Roti
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mark Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology,, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mary V Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Steven Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Teachey
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - William E Evans
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
23
|
Chatwin HV, Cruz Cruz J, Green AL. Pediatric high-grade glioma: moving toward subtype-specific multimodal therapy. FEBS J 2021; 288:6127-6141. [PMID: 33523591 DOI: 10.1111/febs.15739] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
Pediatric high-grade gliomas (pHGG) comprise a deadly, heterogenous category of pediatric gliomas with a clear need for more effective treatment options. Advances in high-throughput molecular techniques have enhanced molecular understanding of these tumors, but outcomes are still poor, and treatments beyond resection and radiation have not yet been clearly established as standard of care. In this review, we first discuss the history of treatment approaches to pHGG to this point. We then review four distinct categories of pHGG, including histone 3-mutant, IDH-mutant, histone 3/IDH-wildtype, and radiation-induced pHGG. We discuss the molecular understanding of each subgroup and targeted treatment options in development. Finally, we look at the development and current status of two novel approaches to pHGG as a whole: localized convection-enhanced chemotherapy delivery and immunotherapy, including checkpoint inhibitors, vaccine therapy, and CAR-T cells. Through this review, we demonstrate the potential for rational, molecularly driven, subtype-specific therapy to be used with other novel approaches in combinations that could meaningfully improve the prognosis in pHGG.
Collapse
Affiliation(s)
- Hannah V Chatwin
- Department of Pediatrics, Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Joselyn Cruz Cruz
- Department of Pediatrics, Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Adam L Green
- Department of Pediatrics, Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado School of Medicine, Aurora, CO, USA.,Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
24
|
Chen M, Zhu Y, Lin Y, Tengwang T, Zhang L. Use of tyrosine kinase inhibitors for paediatric Philadelphia chromosome-positive acute lymphoblastic leukaemia: a systematic review and meta-analysis. BMJ Open 2021; 11:e042814. [PMID: 33468532 PMCID: PMC7817804 DOI: 10.1136/bmjopen-2020-042814] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To investigate the effectiveness and safety of tyrosine kinase inhibitors (TKIs) in the management of paediatric Philadelphia chromosome-positive acute lymphoblastic leukaemia (Ph+ALL). DESIGN A systematic review and meta-analysis. DATA SOURCES Electronic searches were conducted on CENTRAL, MEDLINE, EMBASE, SIOP, ASPHO, ASCO, ASH and four Chinese databases from inception to 8 March 2020. Language of publications was restricted in English and Chinese. ELIGIBILITY CRITERIA Prospective and retrospective comparative studies were included. DATA EXTRACTION AND SYNTHESIS Two authors independently assessed and extracted data. Quality of studies was assessed by the Cochrane Collaboration's tool and Newcastle-Ottawa Scale. Subgroup analysis was performed by comparing different types of TKIs. Quality of evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation approach. RESULTS Two randomised controlled trials (RCTs) and four cohort studies enrolling 536 patients were included. For RCTs, the pooled HR was 0.68 (95% CI 0.26 to 1.78) in overall survival (OS), 0.63 (95% CI 0.28 to 1.42) in event-free survival (EFS), respectively, comparing TKI arm with non-TKI arm for treatment of paediatric Ph+ALL. There was significant difference in OS and EFS between imatinib arm and dasatinib arm (HR 2.26, 95% CI 1.02 to 5.01; HR 2.36; 95% CI 1.27 to 4.39, respectively). For cohort studies, the pooled HR was 0.25 (95% CI 0.14 to 0.47) in OS, 0.25 (95% CI 0.12 to 0.56) in EFS, respectively, comparing TKI arm with non-TKI arm. There was no significance difference in adverse drug reaction between TKI group and without TKI group (risk ratio (RR) 0.82, 95% CI 0.63 to 1.08 in RCT; RR 1.01, 95% CI 0.64 to 1.59 in cohort studies; respectively), and imatinib versus dasatinib (RR 0.97, 95% CI 0.77 to 1.23). The quality of evidence was rated as low for OS, EFS and adverse drug reaction (ADR). CONCLUSIONS The combination of TKIs with chemotherapy is likely to improve the OS and EFS rates in paediatric Ph+ALL, and dasatinib is superior than imatinib. Large sample size and prospective controlled studies are warranted. PROSPERO REGISTRATION NUMBER CRD42018104107.
Collapse
Affiliation(s)
- Min Chen
- Department of Pharmacy/Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Yiping Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
- Department of Pediatric Hematology and Oncology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunzhu Lin
- Department of Pharmacy/Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Tianzi Tengwang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Lingli Zhang
- Department of Pharmacy/Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Carlberg VM, Davies OMT, Brandling-Bennett HA, Leary SES, Huang JT, Coughlin CC, Gupta D. Cutaneous reactions to pediatric cancer treatment part II: Targeted therapy. Pediatr Dermatol 2021; 38:18-30. [PMID: 33378085 DOI: 10.1111/pde.14495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer remains a leading cause of morbidity and mortality among children. Targeted therapies may improve survivorship; however, unique side-effect profiles have also emerged with these novel therapies. Changes in hair, skin, and nails-termed dermatologic adverse events (AEs)-are among the most common sequelae and may result in interruption or discontinuation of therapy. Though dermatologic AEs have been detailed in adults, these findings are not well described in the pediatric population. We reviewed the literature to characterize dermatologic AEs to anticancer targeted therapies available as of July 2020 and summarized the spectrum of clinical findings as well as treatment recommendations for children. Dermatologic AEs are among the most common AEs reported in pediatric patients receiving targeted therapy, but morphologic and histologic descriptions are often lacking in current publications. Pediatric dermatologists are uniquely poised to recognize specific morphology of dermatologic AEs and make recommendations for prevention and treatment that may improve quality of life and enable ongoing cancer therapy.
Collapse
Affiliation(s)
- Valerie M Carlberg
- Children's Wisconsin, Milwaukee, WI, USA.,Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | - Sarah E S Leary
- Seattle Children's Hospital, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - Jennifer T Huang
- Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Carrie C Coughlin
- St Louis Children's Hospital, St. Louis, MO, USA.,Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Deepti Gupta
- Seattle Children's Hospital, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Elsayed HRH, El-Nablaway M, Othman BH, Abdalla AM, El Nashar EM, Abd-Elmonem MM, El-Gamal R. Can Dasatinib Ameliorate the Hepatic changes, Induced by Long Term Western Diet, in Mice? Ann Anat 2020; 234:151626. [PMID: 33144268 DOI: 10.1016/j.aanat.2020.151626] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/20/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a worldwide disease that progresses into steatohepatitis (NASH) that has no current effective treatment. This study aimed, for the first time, to investigate the effect of Dasatinib; a tyrosine kinase inhibitor showing anti-PDGFR activity with a macrophage modulating efficacy, on NASH. METHODS NASH was induced, in C57BL/6 mice by western diet (WD). Control groups received either DMSO or Dasatinib. After 12 weeks, WD-fed mice received DMSO, Dasatinib (4 mg/kg) or Dasatinib (8 mg/kg) once daily, for four weeks. Serum was examined for ALT and lipid profile. Immunohistochemical staining for SREBP1 (lipogenesis marker), iNOS, arginase-1, CD68, CD163 (macrophage polarization markers), TGF-β (fibrosis marker) and ASMA (a marker for activated hepatic stellate cell), hepatic mRNA expression for SREBP-1, iNOS, arginase-1, TGF-β and PDGFA genes; and western blotting for phosphorylated PDGFR α and β, SREBP1, iNOS, arginase-1, IL1α, COX2, TGF-β and ASMA were performed. Liver sections were stained also for H & E, Oil red O and Sirius red. RESULTS Dasatinib could ameliorate the WD-induced disturbance of serum ALT, lipid profile and significantly reduced hepatic expression of PDGFA, phosphorylated PDGFR α and β, IL1α, COX2, SREBP-1, iNOS, CD68, TGF-β and ASMA but increased expression for arginase-1 and CD163 (M2 macrophage markers). Moreover, Dasatinib reduced the steatosis, inflammation, hepatocellular ballooning, hepatic fibrosis and the high NAFLD activity scoring induced by WD. CONCLUSION Dasatinib can prevent the progression of WD-induced NASH by attenuating lipogenesis, and inducing M2 macrophage polarization with antifibrotic activity.
Collapse
Affiliation(s)
| | - Mohammad El-Nablaway
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Egypt
| | - Basma H Othman
- Mansoura Experimental Research Center, Faculty of Medicine, Mansoura University, Egypt
| | - Asim Mohammed Abdalla
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Eman Mohammad El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia; Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | | | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
27
|
Masters JC, Barry E, Knight B. Population Pharmacokinetics of Gemtuzumab Ozogamicin in Pediatric Patients with Relapsed or Refractory Acute Myeloid Leukemia. Clin Pharmacokinet 2020; 58:271-282. [PMID: 30022367 DOI: 10.1007/s40262-018-0694-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND OBJECTIVE To date, the population pharmacokinetics (popPK) of gemtuzumab ozogamicin (GO), a CD33-directed antibody-drug conjugate consisting of hP67.6 antibody linked to N-acetyl gamma calicheamicin used in the treatment of acute myeloid leukemia (AML), has not been characterized in pediatric patients. This report describes the popPK of GO following intravenous administration in 29 pediatric patients aged ≤ 17 years with relapsed or refractory AML who were enrolled in the 0903A1-102-US phase I/II study. METHODS The pharmacokinetics (PK) of GO, as represented by total hP67.6 antibody, were described by a two-compartment model with two clearance components: a linear clearance (CL1) and time-dependent clearance that includes a decay coefficient. The PK of unconjugated calicheamicin (UC; payload) were described by a two-compartment model with CL1 and an input rate of formation based on antibody rate of elimination. Allometric scaling was included in both models, with baseline body weight as a fixed effect on CL1 and central volume. RESULTS AND CONCLUSIONS PK parameters for hP67.6 and UC were not significantly affected by any of the available demographic factors and safety laboratory values tested as covariates (except baseline body weight). Simulations to compare GO dosing regimens (6, 7.5, and 9 mg/m2 on days 1 and 15 versus, 3 mg/m2 fractionated dosing on days 1, 4, and 7) were performed, showing that total antibody and UC trough concentrations were maintained at higher concentrations during treatment following the more frequent dosing than following the original regimen. STUDY IDENTIFIER 0903A1-102-US.
Collapse
Affiliation(s)
- Joanna C Masters
- Clinical Pharmacology, Oncology, Global Product Development, Pfizer Inc, 10555 Science Center Drive, San Diego, CA, 92121, USA.
| | - Elly Barry
- Pfizer Global Product Development Oncology, 300 Technology Square, Suite 302, Cambridge, MA, 02139-3520, USA
| | - Beverly Knight
- Clinical Pharmacology, Oncology, Global Product Development, Pfizer Inc, 10555 Science Center Drive, San Diego, CA, 92121, USA
| |
Collapse
|
28
|
Phase 2 study of nilotinib in pediatric patients with Philadelphia chromosome-positive chronic myeloid leukemia. Blood 2020; 134:2036-2045. [PMID: 31511239 DOI: 10.1182/blood.2019000069] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/17/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic myeloid leukemia (CML) is rare in children and accounts for ≤15% of all myeloid leukemia cases. When we initiated this study with nilotinib, imatinib was the only tyrosine kinase inhibitor indicated for pediatric patients with Philadelphia chromosome-positive (Ph+) CML in chronic phase (CP); alternative treatment options were needed, particularly for patients who developed resistance or intolerance (R/I) to imatinib. This phase 2 study enrolled pediatric patients with either Ph+ CML-CP R/I to imatinib or dasatinib or newly diagnosed Ph+ CML-CP. Data presented are from analyses with minimum follow-up of up to 24 cycles (1 cycle is 28 days). Fifty-nine patients with Ph+ CML-CP were enrolled, and 58 were treated (R/I, n = 33; newly diagnosed, n = 25). Major molecular response (MMR) rate at cycle 6 in the R/I cohort was 39.4% (primary end point); 57.6% of patients achieved or maintained MMR and 81.8% achieved or maintained complete cytogenetic response (CCyR) by 24 cycles. In patients with newly diagnosed disease, rates of MMR by cycle 12 and CCyR at cycle 12 were 64.0% each (primary end points); by cycle 24, cumulative MMR and CCyR rates were 68.0% and 84.0%, respectively. The safety profile of nilotinib in pediatric patients was generally comparable with the known safety profile in adults, although cardiovascular events were not observed in this study, and hepatic laboratory abnormalities were more frequent; no new safety signals were identified. In summary, nilotinib demonstrated efficacy and a manageable safety profile in pediatric patients with Ph+ CML-CP. This trial was registered at www.clinicaltrials.gov as #NCT01844765.
Collapse
|
29
|
Bochennek K, Luckowitsch M, Lehrnbecher T. Recent advances and future directions in the management of the immunocompromised host. Semin Oncol 2020; 47:40-47. [DOI: 10.1053/j.seminoncol.2020.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
|
30
|
Schott C, Shah AT, Sweet-Cordero EA. Genomic Complexity of Osteosarcoma and Its Implication for Preclinical and Clinical Targeted Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:1-19. [PMID: 32767231 DOI: 10.1007/978-3-030-43085-6_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Osteosarcoma is a genomically complex disease characterized by few recurrent single-nucleotide mutations or in-frame fusions. In contrast, structural alterations, including copy number changes, chromothripsis, kataegis, loss of heterozygosity (LOH), and other large-scale genomic alterations, are frequent and widespread across the osteosarcoma genome. These observed structural alterations lead to activation of oncogenes and loss of tumor suppressors which together contribute to oncogenesis. To date, few targeted therapies for osteosarcoma have been identified. It is likely that effectiveness of targeted therapies will vary greatly in subsets of tumors with distinct key driver events. Model systems which can recapitulate the genetic heterogeneity of this disease are needed to test this hypothesis. One possible approach is to use patient-derived xenograft (PDX) models characterized with regards to their similarity to the human tumor samples from which they were derived. Here we review evidence pointing to the genomic complexity of osteosarcoma and how this is reflected in available model systems. We also review the current state of preclinical testing for targeted therapies using these models.
Collapse
Affiliation(s)
- Courtney Schott
- Department of Pediatrics, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Avanthi Tayi Shah
- Department of Pediatrics, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA
| | - E Alejandro Sweet-Cordero
- Department of Pediatrics, Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
31
|
Receptor Tyrosine Kinases in Osteosarcoma: 2019 Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:141-155. [PMID: 32767239 DOI: 10.1007/978-3-030-43085-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary conclusions of our 2014 contribution to this series were as follows: Multiple receptor tyrosine kinases (RTKs) likely contribute to aggressive phenotypes in osteosarcoma and, therefore, inhibition of multiple RTKs is likely necessary for successful clinical outcomes. Inhibition of multiple RTKs may also be useful to overcome resistance to inhibitors of individual RTKs as well as resistance to conventional chemotherapies. Different combinations of RTKs are likely important in individual patients. AXL, EPHB2, FGFR2, IGF1R, and RET were identified as promising therapeutic targets by our in vitro phosphoproteomic/siRNA screen of 42 RTKs in the highly metastatic LM7 and 143B human osteosarcoma cell lines. This chapter is intended to provide an update on these topics as well as the large number of osteosarcoma clinical studies of inhibitors of multiple tyrosine kinases (multi-TKIs) that were recently published.
Collapse
|
32
|
Abstract
Tyrosine kinase inhibitors have recently become an essential tool in management of chronic myeloid leukaemia (CML). Dasatinib, a representative of those drugs, acts by inhibiting key proteins included in CML development, predominantly Bcr-Abl and Src. Its advantage is that it shows activity in many cases where other agents bring no improvement due to resistance. Pharmacokinetics of dasatinib has specific characteristics that may play an important role in achieving sufficient exposure in patients. Therefore, the key pharmacokinetic properties are summarized in this report. For example, dasatinib absorption is significantly influenced by gastric pH and its modulation can be a source of serious interactions, as well as simultaneous administration of drugs affecting cytochrome P450.
Collapse
Affiliation(s)
- Jana Hořínková
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
33
|
Wei J, Ma L, Li C, Pierson CR, Finlay JL, Lin J. Targeting Upstream Kinases of STAT3 in Human Medulloblastoma Cells. Curr Cancer Drug Targets 2019; 19:571-582. [PMID: 30332965 PMCID: PMC6533162 DOI: 10.2174/1568009618666181016165604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 08/21/2018] [Accepted: 09/28/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Medulloblastoma is the most common malignant brain tumor in children. Despite improvement in overall survival rate, it still lacks an effective targeted treatment strategy. The Janus family of cytoplasmic tyrosine kinases (JAKs) and Src kinases, upstream protein kinases of signal transducer and activator of transcription 3 (STAT3), play important roles in medulloblastoma pathogenesis and therefore represent potential therapeutic targets. METHODS In this report, we examined the inhibitory efficacy of the JAK1/2 inhibitor, ruxolitinib, the JAK3 inhibitor, tofacitinib and two Src inhibitors, KX2-391 and dasatinib. RESULTS These small molecule drugs significantly reduce cell viability and inhibit cell migration and colony formation in human medulloblastoma cells in vitro. Src inhibitors have more potent efficacy than JAK inhibitors in inhibiting medulloblastoma cell migration ability. The Src inhibitors can inhibit both phosphorylation of STAT3 and Src while JAK inhibitors reduce JAK/STAT3 phosphorylation. We also investigated the combined effect of the Src inhibitor, dasatinib with cisplatin. The results show that dasatinib exerts synergistic effects with cisplatin in human medulloblastoma cells through the inhibition of STAT3 and Src. CONCLUSION Our results suggest that the small molecule inhibitors of STAT3 upstream kinases, ruxolitinib, tofacitinib, KX2-391, and dasatinib could be novel and attractive candidate drugs for the treatment of human medulloblastoma.
Collapse
Affiliation(s)
- Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Ling Ma
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Chenglong Li
- College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Christopher R. Pierson
- Department of Pathology and Laboratory Medicine, Nationwide Children ‘s Hospital, The Department of Pathology and Department of Biomedical Education & Anatomy, The College of Medicine, The Ohio State University, Columbus,OH 43205, USA
| | - Jonathan L. Finlay
- Division of Hematology, Oncology and BMT, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43205, USA
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
34
|
Kurosawa H, Tanizawa A, Muramatsu H, Tono C, Watanabe A, Shima H, Ito M, Yuza Y, Hamamoto K, Hotta N, Okada M, Saito AM, Manabe A, Mizutani S, Adachi S, Horibe K, Ishii E, Shimada H. Sequential use of second-generation tyrosine kinase inhibitors following imatinib therapy in pediatric chronic myeloid leukemia: A report from the Japanese Pediatric Leukemia/Lymphoma Study Group. Pediatr Blood Cancer 2018; 65:e27368. [PMID: 30084127 DOI: 10.1002/pbc.27368] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/26/2018] [Accepted: 06/29/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND The details of the sequential use of imatinib for first-line treatment followed by second-generation tyrosine kinase inhibitors (2G-TKIs) for pediatric chronic myeloid leukemia (CML) are still unknown. This study analyzed clinical responses and adverse effects of the use of 2G-TKIs following imatinib in pediatric chronic phase (CP)-CML. PROCEDURES The Japanese Pediatric Leukemia/Lymphoma Study Group conducted a retrospective study of patients with newly diagnosed CML from 1996 to 2011. A total of 152 cases that received imatinib as first-line therapy were analyzed. RESULTS Excluding 46 cases treated with hematopoietic stem cell transplantation before nilotinib and dasatinib became available, 31 of 106 patients changed to 2G-TKIs. The primary reason for changing from imatinib was poor response, followed by intolerance, with the main reason for the latter being musculoskeletal events. Switches from imatinib to 2G-TKIs with intolerance occurred significantly earlier than switches with poor response. Sixteen and 15 patients were treated with nilotinib and dasatinib, respectively, following imatinib therapy. After switching to 2G-TKIs, the response status improved in 63% of evaluable patients. The adverse effect profiles of nilotinib and dasatinib tended to be different, with hyperbilirubinemia observed in 33% of nilotinib-treated patients, but in none of the cases with dasatinib. CONCLUSION This retrospective study represents the first series of children and adolescents in whom sequential use of imatinib followed by 2G-TKIs was reported. These data provide useful insights into the selection of 2G-TKIs as first-line treatment for children and adolescents with CP-CML.
Collapse
Affiliation(s)
- Hidemitsu Kurosawa
- Department of Pediatrics, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Akihiko Tanizawa
- Department of Human Resource Development for Cancer, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chikako Tono
- Department of Pediatrics, Iwate Prefectural Chubu Hospital, Iwate, Japan
| | - Akihiro Watanabe
- Department of Pediatrics, Niigata Cancer Center Hospital, Niigata, Japan
| | - Haruko Shima
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.,Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Masaki Ito
- Department of Pediatrics, Soma General Hospital, Fukushima, Japan
| | - Yuki Yuza
- Department of Hematology and Oncology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Kazuko Hamamoto
- Department of Pediatrics, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Noriko Hotta
- Department of Pediatrics, Japan Community Healthcare Organization Tokuyama Central Hospital, Tokuyama, Japan
| | - Masahiko Okada
- Department of Pediatrics, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Akiko Moriya Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Atsushi Manabe
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Shuki Mizutani
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Eiichi Ishii
- Department of Pediatrics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroyuki Shimada
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.,Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
35
|
Mashimo K, Tsubaki M, Takeda T, Asano R, Jinushi M, Imano M, Satou T, Sakaguchi K, Nishida S. RANKL-induced c-Src activation contributes to conventional anti-cancer drug resistance and dasatinib overcomes this resistance in RANK-expressing multiple myeloma cells. Clin Exp Med 2018; 19:133-141. [DOI: 10.1007/s10238-018-0531-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/27/2018] [Indexed: 12/23/2022]
|
36
|
Broniscer A, Jia S, Mandrell B, Hamideh D, Huang J, Onar-Thomas A, Gajjar A, Raimondi SC, Tatevossian RG, Stewart CF. Phase 1 trial, pharmacokinetics, and pharmacodynamics of dasatinib combined with crizotinib in children with recurrent or progressive high-grade and diffuse intrinsic pontine glioma. Pediatr Blood Cancer 2018; 65. [PMID: 29512900 PMCID: PMC5980705 DOI: 10.1002/pbc.27035] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Progressive/recurrent high-grade and diffuse intrinsic pontine gliomas (DIPGs) are fatal. Treatments targeting molecular pathways critical for these cancers are needed. METHODS We conducted a phase 1 study (rolling-six design) to establish the safety and maximum tolerated dose (MTD) of dasatinib, an oral platelet-derived growth factor receptor A (PDGFRA) inhibitor, and crizotinib, an oral c-Met inhibitor, in such patients. Pharmacokinetics of both agents were performed. Biomarkers of cellular pathway activation in peripheral-blood mononuclear cells (PBMC) were evaluated before and after administration of dasatinib. PDGFRA and MET amplification, and PDGFRA mutations were studied in tumor samples. RESULTS Twenty-five patients were enrolled in this study (median age: 11.9 years). Eleven patients had DIPG. Glioblastoma accounted for 40% of cases. Dasatinib at 50 mg/m2 and crizotinib at 130 mg/m2 or 100 mg/m2 were poorly tolerated when administered twice daily. Drug administration was then switched to once daily. Dasatinib administered at 50 mg/m2 and crizotinib at 215 mg/m2 once daily was the MTD. Dose-limiting toxicities consisted of diarrhea, fatigue, proteinuria, hyponatremia, rash, and grade 4 neutropenia. Only two patients received therapy for at least 6 months. No objective radiologic responses were observed. Pharmacokinetics of dasatinib and crizotinib were comparable to previous studies. A statistically significant decrease in the ratio of p-AKT/total AKT in PBMC occurred after dasatinib administration. PDGFRA and MET amplification were found in four and two cases, respectively. Only one of 10 tumors harbored a PDGFRA mutation. CONCLUSIONS This drug combination was poorly tolerated and its activity was minimal. We do not recommend further testing of this combination in children.
Collapse
Affiliation(s)
- Alberto Broniscer
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennesssee
| | - Sujuan Jia
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Belinda Mandrell
- Department of Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Dima Hamideh
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Jie Huang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Amar Gajjar
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennesssee
| | - Susana C. Raimondi
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Ruth G. Tatevossian
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Clinton F. Stewart
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| |
Collapse
|
37
|
Visalli T, Bower N, Kokate T, Andrews PA. Lack of value of juvenile animal toxicity studies for supporting the safety of pediatric oncology phase I trials. Regul Toxicol Pharmacol 2018; 96:167-177. [PMID: 29763632 DOI: 10.1016/j.yrtph.2018.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/07/2018] [Accepted: 05/11/2018] [Indexed: 02/04/2023]
Abstract
Toxicity studies in juvenile animals (JAS) are sometimes performed to support clinical trials in pediatric oncology patients, and there are differing conclusions on the value of JAS for pediatric drug development. This manuscript provides a review of the pediatric clinical data for 25 molecularly-targeted and 4 biologic anticancer therapeutics. Other publications that evaluated the value of JAS in pediatric drug development focus on differences in toxicity between juvenile animals and adult animals. The present paper examines pediatric-specific clinical findings to focus on dose setting in pediatric oncology patients and safety monitoring in terms of the potential value of JAS. Our assessment demonstrates that pediatric starting doses were safe for all 29 therapeutics examined in that no life-threatening toxicities occurred in the first cohort, and overall the ratio of the pediatric maximum tolerated dose (MTD) to the recommended adult dose was close to 1. In addition, the 4 serious adverse events (SAEs) that weren't detectable with standard monitoring plans for pediatric oncology trials would not have been detectable in a standard JAS. This review demonstrates that safe starting doses in pediatric oncology patients for these therapeutics could have been solely based on adult doses without any knowledge of findings in JAS.
Collapse
Affiliation(s)
- Thomas Visalli
- Eisai Inc., Global Nonclinical Regulatory Affairs, 155 Tice Boulevard, Woodcliff Lake, NJ 07677, United States.
| | - Nancy Bower
- Eisai Inc., Global Nonclinical Regulatory Affairs, 155 Tice Boulevard, Woodcliff Lake, NJ 07677, United States
| | - Tushar Kokate
- Eisai Inc., Global Nonclinical Regulatory Affairs, 155 Tice Boulevard, Woodcliff Lake, NJ 07677, United States
| | - Paul A Andrews
- Eisai Inc., Global Nonclinical Regulatory Affairs, 155 Tice Boulevard, Woodcliff Lake, NJ 07677, United States
| |
Collapse
|
38
|
Gore L, Kearns PR, de Martino ML, Lee, De Souza CA, Bertrand Y, Hijiya N, Stork LC, Chung NG, Cardos RC, Saikia T, Fagioli F, Seo JJ, Landman-Parker J, Lancaster D, Place AE, Rabin KR, Sacchi M, Swanink R, Zwaan CM. Dasatinib in Pediatric Patients With Chronic Myeloid Leukemia in Chronic Phase: Results From a Phase II Trial. J Clin Oncol 2018; 36:1330-1338. [PMID: 29498925 DOI: 10.1200/jco.2017.75.9597] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Safe, effective treatments are needed for pediatric patients with chronic myeloid leukemia in chronic phase (CML-CP). Dasatinib is approved for treatment of adults and children with CML-CP. A phase I study determined suitable dosing for children with Philadelphia chromosome-positive (Ph+) leukemias. Methods CA180-226/NCT00777036 is a phase II, open-label, nonrandomized prospective trial of patients < 18 years of age receiving dasatinib. There are three cohorts: (1) imatinib-resistant/intolerant CML-CP, (2) imatinib-resistant/intolerant CML in accelerated/blast phase or Ph+ acute lymphoblastic leukemia (n = 17), and (3) newly diagnosed CML-CP treated with tablets or powder for oral suspension. Major cytogenetic response > 30% for imatinib-resistant/intolerant patients and complete cytogenetic response (CCyR) > 55% for newly diagnosed patients were of clinical interest. Results Of 113 patients with CML-CP, 14 (48%) who were imatinib-resistant/intolerant and 61 (73%) who were newly diagnosed remained on treatment at time of analysis. Major cytogenetic response > 30% was reached by 3 months in the imatinib-resistant/intolerant group and CCyR > 55% was reached by 6 months in the newly diagnosed CML-CP group. CCyR and major molecular response by 12 months, respectively, were 76% and 41% in the imatinib-resistant/intolerant group and 92% and 52% in newly diagnosed CML-CP group. Progression-free survival by 48 months was 78% and 93% in the imatinib-resistant/intolerant and newly diagnosed CML-CP groups, respectively. No dasatinib-related pleural or pericardial effusion, pulmonary edema, or pulmonary arterial hypertension were reported. Bone growth and development events were reported in 4% of patients. Conclusion In the largest prospective trial to date in children with CML-CP, we demonstrate that dasatinib is a safe, effective treatment of pediatric CML-CP. Target responses to first- or second-line dasatinib were met early, and deep molecular responses were observed. Safety of dasatinib in pediatric patients was similar to that observed in adults; however, no cases of pleural or pericardial effusion or pulmonary arterial hypertension were reported.
Collapse
Affiliation(s)
- Lia Gore
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Pamela R Kearns
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Maria Lucia de Martino
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Lee
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Carmino Antonio De Souza
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Yves Bertrand
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Nobuko Hijiya
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Linda C Stork
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Nack-Gyun Chung
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Rocio Cardenas Cardos
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Tapan Saikia
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Franca Fagioli
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Jong Jin Seo
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Judith Landman-Parker
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Donna Lancaster
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Andrew E Place
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Karen R Rabin
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Mariana Sacchi
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - Rene Swanink
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| | - C Michel Zwaan
- Lia Gore, University of Colorado School of Medicine/Children's Hospital Colorado, Aurora, CO; Pamela R. Kearns, University of Birmingham, Birmingham, West Midlands; Donna Lancaster, Royal Marsden Hospital, Sutton, Surrey, United Kingdom; Maria Lucia de Martino Lee, Support Group for Children and Adolescents with Cancer; Carmino Antonio De Souza, University of Campinas, São Paulo, Brazil; Yves Bertrand, L'Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France; Nobuko Hijiya, Ann and Robert H. Lurie Children's Hospital of Chicago, and Northwestern University Feinberg School of Medicine, Chicago, IL; Linda C. Stork, Oregon Health & Science University, Portland, OR; Nack-Gyun Chung, The Catholic University of Korea, Seoul St. Mary's Hospital; Jong Jin Seo, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Republic of Korea; Rocio Cardenas Cardos, Instituto Nacional De Pediatria, Mexico City, Mexico; Tapan Saikia, Prince Aly Khan Hospital, Mumbai, India; Franca Fagioli, Regina Margherita Hospital, Turin, Italy; Judith Landman-Parker, Hôpital Enfants Armand-Trousseau, Paris, France; Andrew E. Place, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Karen R. Rabin, Texas Children's Cancer Center, and Baylor College of Medicine, Houston, TX; Mariana Sacchi and Rene Swanink, Bristol-Myers Squibb, Princeton, NJ; C. Michel Zwaan, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands; Pamela R. Kearns and C. Michel Zwaan, Innovative Therapies for Children with Cancer Consortium, European Union
| |
Collapse
|
39
|
Robison NJ, Yeo KK, Berliner AP, Malvar J, Sheard MA, Margol AS, Seeger RC, Rushing T, Finlay JL, Sposto R, Dhall G. Phase I trial of dasatinib, lenalidomide, and temozolomide in children with relapsed or refractory central nervous system tumors. J Neurooncol 2018; 138:199-207. [PMID: 29427149 DOI: 10.1007/s11060-018-2791-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/01/2018] [Indexed: 01/12/2023]
Abstract
Single agent studies targeting the tumor microenvironment in central nervous system (CNS) tumors have largely been disappointing. Combination therapies targeting various pathways and cell types may be a more effective strategy. In this phase I study, we evaluated the combination of dasatinib, lenalidomide, and temozolomide in children with relapsed or refractory primary CNS tumors. Patients 1-21 years old with relapsed or refractory CNS tumors were eligible. Starting doses of dasatinib and lenalidomide were 65 mg/m2/dose twice daily and 55 mg/m2 once daily, respectively, while temozolomide was constant at 75 mg/m2 daily. The study followed a 3 + 3 phase I design, with a 4-week dose-limiting toxicity (DLT) evaluation period. Serial peripheral blood lymphocyte subsets were evaluated in consenting patients. Fifteen patients were enrolled and thirteen were DLT-evaluable. DLTs occurred in 5 patients, including somnolence and confusion (1 patient), hypokalemia (1 patient) and thrombocytopenia (3 patients). The maximum tolerated dose for the combination was dasatinib 65 mg/m2 twice daily, lenalidomide 40 mg/m2 daily, and temozolomide 75 mg/m2 daily, for 21 days followed by 7 days rest in repeating 28-day cycles. Transient increases in natural killer effector cells and cytotoxic T-cells were seen after 1 week of treatment. One out of six response-evaluable patients showed a partial response. The combination was feasible and relatively well tolerated in this heavily pre-treated population. The most common toxicities were hematologic. Preliminary evidence of clinical benefit was seen.
Collapse
Affiliation(s)
- Nathan J Robison
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA. .,University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| | - Kee Kiat Yeo
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Adrian P Berliner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Jemily Malvar
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Michael A Sheard
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Ashley S Margol
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Robert C Seeger
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Teresa Rushing
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Jonathan L Finlay
- Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Richard Sposto
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Girish Dhall
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, 4650 Sunset Boulevard, MS #54, Los Angeles, CA, 90027, USA.,University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
40
|
Kimura Y, Chisaki Y, Saki T, Matsumura C, Motohashi H, Onoue M, Yano Y. Prediction of Apparent Oral Clearance of Small-Molecule Inhibitors in Pediatric Patients. J Pharm Sci 2017; 107:949-956. [PMID: 29133236 DOI: 10.1016/j.xphs.2017.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/14/2017] [Accepted: 11/01/2017] [Indexed: 11/15/2022]
Abstract
The purpose of this study was to build regression models for the prediction of apparent oral clearance (CL/F) for small-molecule inhibitors in the pediatric population using data obtained from adults. Two approaches were taken; a simple allometric regression model which considers no interdrug or interindividual variability and an allometric regression model with mixed-effects modeling where some variability parameters are included in the model. Average CL/F values were obtained for 15 drugs at various dosages from 31 literatures (a total of 139 data sets) conducted in adults and for 15 drugs from 26 literatures (62 data sets) conducted in children. Data were randomly separated into the "modeling" or "validation" data set, and the 2 allometric regression models were applied to the modeling data set. The predictive ability of the models was examined by comparing the observed and model-predicted CL/F in children using the validation data set. The percentage root mean square error was 17.2% and 26.3% in the simple allometric regression model and the allometric regression model with mixed-effects modeling, respectively. The predictive ability of the 2 models seems acceptable, suggesting that they could be useful for predicting the CL/F of new small-molecule inhibitors and for determining adequate doses in clinical pharmacotherapy for children.
Collapse
Affiliation(s)
- Yoshihiko Kimura
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan; Department of Pharmacy, Kitano Hospital, The Tazuke Kofukai Medical Research Institute, Kita-ku, Osaka, 530-8480, Japan
| | - Yugo Chisaki
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Tomohiko Saki
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Chikako Matsumura
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Hideyuki Motohashi
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Masahide Onoue
- Department of Pharmacy, Kitano Hospital, The Tazuke Kofukai Medical Research Institute, Kita-ku, Osaka, 530-8480, Japan
| | - Yoshitaka Yano
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| |
Collapse
|
41
|
Dorris K, Liu C, Li D, Hummel TR, Wang X, Perentesis J, Kim MO, Fouladi M. A comparison of safety and efficacy of cytotoxic versus molecularly targeted drugs in pediatric phase I solid tumor oncology trials. Pediatr Blood Cancer 2017; 64. [PMID: 27654490 DOI: 10.1002/pbc.26258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/19/2016] [Accepted: 08/10/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND Prior reviews of phase I pediatric oncology trials involving primarily cytotoxic agents have reported objective response rates (ORRs) and toxic death rates of 7.9-9.6% and 0.5%, respectively. These data may not reflect safety and efficacy in phase I trials of molecularly targeted (targeted) drugs. METHODS A systematic review of pediatric phase I solid tumor trials published in 1990-2013 was performed. The published reports were evaluated for patient characteristics, toxicity information, and response numbers. RESULTS A total of 143 phase I pediatric clinical trials enrolling 3,896 children involving 53 targeted and 48 cytotoxic drugs were identified. A meta-analysis demonstrated that the ORR is 2.1-fold higher with cytotoxic drugs (0.066 vs. 0.031 per subject; P = 0.007). By contrast, the pooled estimate of the stable disease rate (SDR) is similar for cytotoxic and targeted drugs (0.2 vs. 0.23 per subject; P = 0.27). The pooled estimate of the dose-limiting toxicity rate is 1.8-fold larger with cytotoxic drugs (0.24 vs. 0.13 per subject; P = 0.0003). The hematologic grade 3-4 (G3/4) toxicity rate is 3.6-fold larger with cytotoxic drugs (0.43 vs. 0.12 per treatment course; P = 0.0001); however, the nonhematologic G3/4 toxicities and toxic deaths occur at similar rates for cytotoxic and targeted drugs. CONCLUSIONS In phase I pediatric solid tumor trials, ORRs were significantly higher for cytotoxic versus targeted agents. SDRs were similar in targeted and cytotoxic drug trials. Patients treated with cytotoxic agents were more likely to experience hematologic G3/4 toxicities than those patients receiving targeted drugs.
Collapse
Affiliation(s)
- Kathleen Dorris
- Section of Pediatric Hematology, Oncology, Bone Marrow Transplantation, Children's Hospital Colorado, Aurora, Colorado
| | - Chunyan Liu
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dandan Li
- Consumer Credit Risk Management, Fifth Third Bank, Cincinnati, Ohio
| | - Trent R Hummel
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Xia Wang
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, Ohio
| | - John Perentesis
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Mi-Ok Kim
- Department of Epidemiology and Biostatistics, University of California San Francisco
| | - Maryam Fouladi
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
42
|
Koschmann C, Zamler D, MacKay A, Robinson D, Wu YM, Doherty R, Marini B, Tran D, Garton H, Muraszko K, Robertson P, Leonard M, Zhao L, Bixby D, Peterson L, Camelo-Piragua S, Jones C, Mody R, Lowenstein PR, Castro MG. Characterizing and targeting PDGFRA alterations in pediatric high-grade glioma. Oncotarget 2016; 7:65696-65706. [PMID: 27582545 PMCID: PMC5323185 DOI: 10.18632/oncotarget.11602] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/13/2016] [Indexed: 12/20/2022] Open
Abstract
Pediatric high-grade glioma (HGG, WHO Grade III and IV) is a devastating brain tumor with a median survival of less than two years. PDGFRA is frequently mutated/ amplified in pediatric HGG, but the significance of this finding has not been fully characterized. We hypothesize that alterations of PDGFRA will promote distinct prognostic and treatment implications in pediatric HGG. In order to characterize the impact of PDGFR pathway alterations, we integrated genomic data from pediatric HGG patients (n=290) from multiple pediatric datasets and sequencing platforms. Integration of multiple human datasets showed that PDGFRA mutation, but not amplification, was associated with older age in pediatric HGG (P= <0.0001). In multivariate analysis, PDGFRA mutation was correlated with worse prognosis (P = 0.026), while PDGFRA amplification was not (P = 0.11). By Kaplan-Meier analysis, non-brainstem HGG with PDGFRA amplification carried a worse prognosis than non-brainstem HGG without PDGFRA amplification (P = 0.021). There were no pediatric patients with PDGFRA-amplified HGG that survived longer than two years. Additionally, we performed paired molecular profiling (germline / tumor / primary cell culture) and targeting of an infant thalamic HGG with amplification and outlier increased expression of PDGFRA. Dasatinib inhibited proliferation most effectively. In summary, integration of the largest genomic dataset of pediatric HGG to date, allowed us to highlight that PDGFRA mutation is found in older pediatric patients and that PDGFRA amplification is prognostic in non-brainstem HGG. Future precision-medicine based clinical trials for pediatric patients with PDGFRA-altered HGG should consider the optimized delivery of dasatinib.
Collapse
Affiliation(s)
- Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Daniel Zamler
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Alan MacKay
- Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, SM2 5NG, UK
| | - Dan Robinson
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yi-Mi Wu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Robert Doherty
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Bernard Marini
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Dustin Tran
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Hugh Garton
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Karin Muraszko
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Patricia Robertson
- Department of Pediatrics, Division of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Marcia Leonard
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Dale Bixby
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Luke Peterson
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Sandra Camelo-Piragua
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Chris Jones
- Divisions of Molecular Pathology and Cancer Therapeutics, Institute of Cancer Research, London, SM2 5NG, UK
| | - Rajen Mody
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
43
|
Heymann MF, Brown HK, Heymann D. Drugs in early clinical development for the treatment of osteosarcoma. Expert Opin Investig Drugs 2016; 25:1265-1280. [DOI: 10.1080/13543784.2016.1237503] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Marie-Françoise Heymann
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, Faculty of Medicine, University of Nantes, Nantes, France
- Nantes University Hospital, Nantes, France
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| | - Hannah K. Brown
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| | - Dominique Heymann
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
- INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, Faculty of Medicine, University of Nantes, Nantes, France
- Nantes University Hospital, Nantes, France
- European Associated Laboratory, Sarcoma Research Unit, Medical School, INSERM-University of Sheffield, Sheffield, UK
| |
Collapse
|
44
|
Outcomes of Patients With Relapsed Hepatoblastoma Enrolled on Children's Oncology Group (COG) Phase I and II Studies. J Pediatr Hematol Oncol 2016; 38:187-90. [PMID: 26583620 DOI: 10.1097/mph.0000000000000474] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Data are limited regarding outcomes of patients treated for relapsed hepatoblastoma. We reviewed enrollment patterns and outcomes of patients with hepatoblastoma on Children's Oncology Group (COG) phase I/II studies. The medical literature was searched for reports of COG phase I/II studies using PUBMED as well as an inventory from the COG publications office searching manuscripts published from 2000 to 2014. Seventy-one patients with relapsed hepatoblastoma were enrolled on 23 separate COG phase I/II studies. Four studies collected α-fetoprotein (AFP) data, but none utilized AFP decline in assessing response. Most studies enrolled few patients with relapsed hepatoblastoma: 7 studies enrolled 1 patient, and another 7 studies enrolled 2 patients each. Only 9 studies enrolled 3 or more patients with relapsed hepatoblastoma. Four responses were reported. Dedicated strata and/or focus on 1 or 2 studies with compelling biological or clinical rationale for hepatoblastoma may improve accrual (and statistical significance of response data) of patients with relapsed hepatoblastoma. Prospective study of AFP decline versus RECIST response could help determine the optimal method of assessing response to identify potentially beneficial treatments in hepatoblastoma.
Collapse
|
45
|
Tanizawa A. Optimal management for pediatric chronic myeloid leukemia. Pediatr Int 2016; 58:171-9. [PMID: 26646444 DOI: 10.1111/ped.12876] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 11/19/2015] [Accepted: 12/01/2015] [Indexed: 12/11/2022]
Abstract
Chronic myeloid leukemia (CML) is rare among childhood leukemias. Its incidence increases with age, from 0.09/100 000 at ≤15 years old to 7.88/100 000 at ≥75 years old. There are several biological and clinical differences between pediatric and adult CML. Markedly increased leukocyte count and a higher incidence of splenomegaly are characteristic features at diagnosis in pediatric patients. The therapeutic approach to CML has changed since the introduction of the tyrosine kinase inhibitor (TKI) imatinib, followed by dasatinib and nilotinib. Given the efficacy of TKI in adult CML, TKI are regarded as the established first-line treatment in adult patients. In 2011, a prospective phase IV study in pediatric patients showed the excellent efficacy and safety of imatinib. Imatinib is also accepted as a first-line option for childhood chronic phase CML. Although the efficacy of dasatinib and nilotinib reported in adult studies seems very attractive for pediatric patients, neither drug has been prospectively investigated in a large pediatric cohort. TKI are designed to inhibit BCR-ABL1 kinase, but they have unfavorable effects, so-called "off-target" complications, such as growth impairment. Long-term morbidity due to TKI is unknown. Furthermore, the adverse effects on growing children have not been clearly elucidated, even though the exposure period to imatinib is relatively short. To establish the standard therapeutic management for pediatric CML, it is important to prospectively confirm the attractive outcomes obtained in adult studies via pediatric clinical trials with a careful monitoring system for TKI-induced adverse effects, especially in growing children.
Collapse
Affiliation(s)
- Akihiko Tanizawa
- Department of Human Resource Development for Cancer, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
46
|
Taga T, Tomizawa D, Takahashi H, Adachi S. Acute myeloid leukemia in children: Current status and future directions. Pediatr Int 2016; 58:71-80. [PMID: 26645706 DOI: 10.1111/ped.12865] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 01/17/2023]
Abstract
Acute myeloid leukemia (AML) accounts for 25% of pediatric leukemia and affects approximately 180 patients annually in Japan. The treatment outcome for pediatric AML has improved through advances in chemotherapy, hematopoietic stem cell transplantation (HSCT), supportive care, and optimal risk stratification. Currently, clinical pediatric AML studies are conducted separately according to the AML subtypes: de novo AML, acute promyelocytic leukemia (APL), and myeloid leukemia with Down syndrome (ML-DS). Children with de novo AML are treated mainly with anthracyclines and cytarabine, in some cases with HSCT, and the overall survival (OS) rate now approaches 70%. Children with APL are treated with an all-trans retinoic acid (ATRA)-combined regimen with an 80-90% OS. Children with ML-DS are treated with a less intensive regimen compared with non-DS patients, and the OS is approximately 80%. HSCT in first remission is restricted to children with high-risk de novo AML only. To further improve outcomes, it will be necessary to combine more accurate risk stratification strategies using molecular genetic analysis with assessment of minimum residual disease, and the introduction of new drugs in international collaborative clinical trials.
Collapse
Affiliation(s)
- Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | | | | |
Collapse
|
47
|
Huguet F. Dasatinib for acute lymphoblastic leukemia. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1098530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Pediatric chronic myeloid leukemia is a unique disease that requires a different approach. Blood 2015; 127:392-9. [PMID: 26511135 DOI: 10.1182/blood-2015-06-648667] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022] Open
Abstract
Chronic myelogenous leukemia (CML) in children is relatively rare. Because of a lack of robust clinical study evidence, management of CML in children is not standardized and often follows guidelines developed for adults. Children and young adults tend to have a more aggressive clinical presentation than older adults, and prognostic scores for adult CML do not apply to children. CML in children has been considered to have the same biology as in adults, but recent data indicate that some genetic differences exist in pediatric and adult CML. Because children with CML may receive tyrosine kinase inhibitor (TKI) therapy for many decades, and are exposed to TKIs during a period of active growth, morbidities in children with CML may be distinct from those in adults and require careful monitoring. Aggressive strategies, such as eradication of CML stem cells with limited duration and intensive regimens of chemotherapy and TKIs, may be more advantageous in children as a way to avoid lifelong exposure to TKIs and their associated adverse effects. Blood and marrow transplantation in pediatric CML is currently indicated only for recurrent progressive disease, and the acute and long-term toxicities of this option should be carefully evaluated against the complications associated with lifelong use of TKIs.
Collapse
|
49
|
Wu KH, Wu HP, Weng T, Peng CT, Chao YH. Dasatinib for a child with Philadelphia chromosome-positive acute lymphoblastic leukemia and persistently elevated minimal residual disease during imatinib therapy. ACTA ACUST UNITED AC 2015; 22:303-6. [PMID: 26300669 DOI: 10.3747/co.22.2719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Imatinib has improved outcomes in patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (all). Minimal residual disease (mrd) is a useful tool for predicting leukemia relapse. However, there is no consensus on how to treat children with elevation of BCR-ABL transcripts but no evidence of hematologic relapse during chemotherapy combined with imatinib. Here, we report the case of a child with Ph+ all who had persistent elevation of mrd, but no evidence of hematologic relapse while receiving imatinib plus intensive chemotherapy. Dasatinib was substituted for imatinib because no suitable donor for allogeneic hematopoietic stem-cell transplantation (hsct) was available. Less-intensive chemotherapy with methotrexate and 6-mercaptopurine was administered concomitantly. No serious adverse events were encountered. With continuous dasatinib combined with chemotherapy, but no allogeneic hsct, our patient reached complete molecular remission and has been in complete molecular remission for more than 13 months. This report is the first about the long-term use of dasatinib in patients with Ph+ all and mrd elevation but hematologic remission during imatinib chemotherapy. In a similar situation, chemotherapy combined with dasatinib instead of allogeneic hsct could be considered to avoid hsct-related mortality and morbidity. Clinical trials are needed.
Collapse
Affiliation(s)
- K H Wu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; ; Department of Pediatrics, Children's Hospital, China Medical University, Taichung, Taiwan
| | - H P Wu
- Department of Pediatrics, Buddhist Tzu-Chi General Hospital, Taichung Branch, Taichung, Taiwan
| | - T Weng
- Department of Pediatrics, Children's Hospital, China Medical University, Taichung, Taiwan
| | - C T Peng
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; ; Department of Pediatrics, Children's Hospital, China Medical University, Taichung, Taiwan; ; Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Y H Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan; ; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
50
|
Belum VR, Washington C, Pratilas CA, Sibaud V, Boralevi F, Lacouture ME. Dermatologic adverse events in pediatric patients receiving targeted anticancer therapies: a pooled analysis. Pediatr Blood Cancer 2015; 62:798-806. [PMID: 25683226 PMCID: PMC4376610 DOI: 10.1002/pbc.25429] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND The dermatologic adverse events (AEs) of various molecularly targeted therapies are well-described in adult cancer patients. Little has been reported on the incidence and clinical presentation of such AEs in pediatric patients with cancer. To address this gap, we analyzed the dermatologic AEs reported across clinical trials of targeted anticancer therapies in pediatric patients. PROCEDURES We conducted an electronic literature search (PubMed, American Society of Clinical Oncology annual meetings' abstracts, ClinicalTrials.gov, NCI's Pediatric Oncology Branch webpage) to identify clinical trials involving targeted anticancer therapies that reported dermatologic AEs in their safety data. Studies were limited to the pediatric population, monotherapy trials (oncology), and English language publications. RESULTS Pooled data from 19 clinical studies investigating 11 targeted anticancer agents (alemtuzumab, rituximab, imatinib, dasatinib, erlotinib, vandetanib, sorafenib, cabozantinib, pazopanib, everolimus, and temsirolimus) were analyzed. The most frequently encountered dermatologic AEs were rash (127/660; 19%), xerosis (18/100; 18%), mucositis (68/402; 17%), and pruritus (12/169; 7%). Other AEs included pigmentary abnormalities of the skin/hair (13%), hair disorders (trichomegaly, hypertrichosis, alopecia, and madarosis; 14%), urticaria (7%), palmoplantar erythrodysesthesia (7%), erythema, acne, purpura, skin fissures, other 'unknown skin changes', exanthem, infection, flushing, telangiectasia, and photosensitivity. CONCLUSION This study describes the dermatologic manifestations of targeted anticancer therapy-related AEs in the pediatric population. Since these AEs are often associated with significant morbidity, it is imperative that pediatric oncologists be familiar with their recognition and management, to avoid unnecessary dose modifications and/or termination, and to prevent impairments in patients' quality of life.
Collapse
Affiliation(s)
| | - Courtney Washington
- Department of Dermatology, Memorial Sloan Kettering Cancer Center, New York, NY USA
- Philadelphia College of Osteopathic Medicine, Suwanee, GA, USA
| | | | - Vincent Sibaud
- Department of Dermatology, Institut Claudius Regaud, Institut Universitaire Cancer Toulouse-oncopole, Toulouse, France
| | - Franck Boralevi
- Unité de Dermatologie Pédiatrique, Hôpital Pellegrin-enfants, Place Amélie Raba-Léon, 33076 Bordeaux Cedex, France
| | - Mario E. Lacouture
- Department of Dermatology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|