1
|
Jerry L, Swan L, Dana N, Balis FM, Greengard E, Huiping X. Population modeling analyses of crizotinib in pediatric patients with ALK-positive advanced cancers. Pediatr Blood Cancer 2024; 71:e31139. [PMID: 38867367 DOI: 10.1002/pbc.31139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Alterations in the ALK (anaplastic lymphoma kinase) gene play a critical role in pathogenesis of anaplastic large cell lymphoma (ALCL). Crizotinib is a small molecule competitive inhibitor of ALK, ROS1, and MET kinases and was approved for pediatric patients with ALK-positive relapsed or refractory, systemic ALCL, and ALK-positive unresectable, recurrent, or refractory inflammatory myofibroblastic tumors (IMT). PROCEDURE Crizotinib data from pediatric patients with relapsed or refractory solid tumors, IMT, or ALCL were included in the analyses. All patients received crizotinib orally at doses ranging from 100 to 365 mg/m2 twice daily (BID). PopPK analyses were conducted to characterize crizotinib disposition in pediatric patients. Exposure-response (ER) safety and antitumor analyses were conducted to characterize relationships between crizotinib dose or exposure with safety and antitumor activity endpoints of interest. RESULTS The population pharmacokinetic (popPK), ER safety, and ER antitumor analysis included 98, 110, and 36 pediatric patients, respectively. A one-compartment pharmacokinetic model with allometric scaling, first-order elimination, and first-order absorption with lag time adequately described the data. Natural log-transformed model-predicted crizotinib AUCss (steady-state area under the concentration-time curve) demonstrated a significant, positive relationship with Grade ≥3 NEUTROPENIA and Any Grade VISION DISORDER. Crizotinib dose demonstrated a positive relationship with objective response rate. CONCLUSIONS No significant differences in PK were identified across a wide range of ages or across tumor types, suggesting body surface area (BSA)-based dosing adequately adjusted for differences in patient size to achieve similar systemic crizotinib exposures across young children and adolescent pediatric patients. None of the myelosuppressive events except Grade ≥3 NEUTROPENIA had significant relationships identified with crizotinib dose or exposure, suggesting crizotinib is a tolerable treatment with less hematological toxicity than traditional chemotherapy regimens for pediatric patients with ALK-mutated cancers. Results from the presented analyses support the pediatric dosing recommendations in the product label.
Collapse
Affiliation(s)
- Li Jerry
- Clinical Pharmacology, Pfizer Inc., New York, New York, USA
| | - Lin Swan
- Clinical Pharmacology, Neurocrine Biosciences, San Diego, California, USA
| | - Nickens Dana
- Clinical Pharmacology, Pfizer Inc., New York, New York, USA
| | - Frank M Balis
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Emily Greengard
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xu Huiping
- Clinical Pharmacology, Pfizer Inc., New York, New York, USA
| |
Collapse
|
2
|
Wang H, Chen H, Cui X, Zhang Y, Zhou J, Chen X. Simultaneous determination of unecritinib (TQ-B3101) and its active metabolite crizotinib in rat plasma by LC-MS/MS:An application to pharmacokinetic studies. J Pharm Biomed Anal 2024; 246:116199. [PMID: 38744200 DOI: 10.1016/j.jpba.2024.116199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/31/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Unecritinib (TQ-B3101) is a selective tyrosine kinase receptor inhibitor. In the study, in vitro metabolic experiments revealed that the hydrolysis of TQ-B3101 was mainly catalyzed by carboxylesterase 2 (CES2), followed by CES1. Next, a sensitive and reliable LC-MS/MS method was established for the simultaneous determination of TQ-B3101 and its metabolite crizotinib in rat plasma. To prevent in vitro hydrolysis of TQ-B3101, sodium fluoride, the CESs inhibitor at a concentration of 2 M, was immediately added after whole blood collection. Plasma samples were extracted by acetonitrile-induced protein precipitation method, and chromatographically separated on a Gemini C18 column (50 mm × 2.0 mm i.d., 5 μm) using gradient elution with a mobile phase of 0.1% formic acid and 5 mmol/L ammonium acetate with 0.1% formic acid. The retention times for TQ-B3101 and crizotinib were 2.61 and 2.38 min, respectively. The analytes were detected with tandem mass spectrometer by positive electrospray ionization, using the ion transitions at m/z 492.3 → 302.3 for TQ-B3101, m/z 450.3 → 260.3 for crizotinib, and m/z 494.0 → 394.3 for imatinib (internal standard). Method validation was conducted in the linear range of 1.00-800 ng/mL for the two analytes. The precision, accuracy and stabilities all met the acceptance criteria. The pharmacokinetic study indicated that TQ-B3101 was rapidly hydrolyzed to crizotinib with the elimination half-life of 1.11 h after a single gavage administration of 27 mg/kg to Sprague-Dawley rats, and the plasma exposure of TQ-B3101 was only 2.98% of that of crizotinib.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China
| | - Huixian Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China
| | - Xinran Cui
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yuchen Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China
| | - Jialan Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China
| | - Xiaoyan Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, PR China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
3
|
Centanni M, Zaher O, Elhad D, Karlsson MO, Friberg LE. Physiologically-based pharmacokinetic models versus allometric scaling for prediction of tyrosine-kinase inhibitor exposure from adults to children. Cancer Chemother Pharmacol 2024; 94:297-310. [PMID: 38782791 PMCID: PMC11390758 DOI: 10.1007/s00280-024-04678-0] [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: 12/05/2023] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Model-based methods can predict pediatric exposure and support initial dose selection. The aim of this study was to evaluate the performance of allometric scaling of population pharmacokinetic (popPK) versus physiologically based pharmacokinetic (PBPK) models in predicting the exposure of tyrosine kinase inhibitors (TKIs) for pediatric patients (≥ 2 years), based on adult data. The drugs imatinib, sunitinib and pazopanib were selected as case studies due to their complex PK profiles including high inter-patient variability, active metabolites, time-varying clearances and non-linear absorption. METHODS Pediatric concentration measurements and adult popPK models were derived from the literature. Adult PBPK models were generated in PK-Sim® using available physicochemical properties, calibrated to adult data when needed. PBPK and popPK models for the pediatric populations were translated from the models for adults and were used to simulate concentration-time profiles that were compared to the observed values. RESULTS Ten pediatric datasets were collected from the literature. While both types of models captured the concentration-time profiles of imatinib, its active metabolite, sunitinib and pazopanib, the PBPK models underestimated sunitinib metabolite concentrations. In contrast, allometrically scaled popPK simulations accurately predicted all concentration-time profiles. Trough concentration (Ctrough) predictions from the popPK model fell within a 2-fold range for all compounds, while 3 out of 5 PBPK predictions exceeded this range for the imatinib and sunitinib metabolite concentrations. CONCLUSION Based on the identified case studies it appears that allometric scaling of popPK models is better suited to predict exposure of TKIs in pediatric patients ≥ 2 years. This advantage may be attributed to the stable enzyme expression patterns from 2 years old onwards, which can be easily related to adult levels through allometric scaling. In some instances, both methods performed comparably. Understanding where discrepancies between the model methods arise, can further inform model development and ultimately support pediatric dose selection.
Collapse
Affiliation(s)
- Maddalena Centanni
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - Omar Zaher
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - David Elhad
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - Mats O Karlsson
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, 751 23, Sweden.
| |
Collapse
|
4
|
Zuckermann M, He C, Andrews J, Bagchi A, Sloan-Henry R, Bianski B, Xie J, Wang Y, Twarog N, Onar-Thomas A, Ernst KJ, Yang L, Li Y, Zhu X, Ocasio JK, Budd KM, Dalton J, Li X, Chepyala D, Zhang J, Xu K, Hover L, Roach JT, Chan KCH, Hofmann N, McKinnon PJ, Pfister SM, Shelat AA, Rankovic Z, Freeman BB, Chiang J, Jones DTW, Tinkle CL, Baker SJ. Capmatinib is an effective treatment for MET-fusion driven pediatric high-grade glioma and synergizes with radiotherapy. Mol Cancer 2024; 23:123. [PMID: 38849845 PMCID: PMC11157767 DOI: 10.1186/s12943-024-02027-6] [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: 11/04/2023] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Pediatric-type diffuse high-grade glioma (pHGG) is the most frequent malignant brain tumor in children and can be subclassified into multiple entities. Fusion genes activating the MET receptor tyrosine kinase often occur in infant-type hemispheric glioma (IHG) but also in other pHGG and are associated with devastating morbidity and mortality. METHODS To identify new treatment options, we established and characterized two novel orthotopic mouse models harboring distinct MET fusions. These included an immunocompetent, murine allograft model and patient-derived orthotopic xenografts (PDOX) from a MET-fusion IHG patient who failed conventional therapy and targeted therapy with cabozantinib. With these models, we analyzed the efficacy and pharmacokinetic properties of three MET inhibitors, capmatinib, crizotinib and cabozantinib, alone or combined with radiotherapy. RESULTS Capmatinib showed superior brain pharmacokinetic properties and greater in vitro and in vivo efficacy than cabozantinib or crizotinib in both models. The PDOX models recapitulated the poor efficacy of cabozantinib experienced by the patient. In contrast, capmatinib extended survival and induced long-term progression-free survival when combined with radiotherapy in two complementary mouse models. Capmatinib treatment increased radiation-induced DNA double-strand breaks and delayed their repair. CONCLUSIONS We comprehensively investigated the combination of MET inhibition and radiotherapy as a novel treatment option for MET-driven pHGG. Our seminal preclinical data package includes pharmacokinetic characterization, recapitulation of clinical outcomes, coinciding results from multiple complementing in vivo studies, and insights into molecular mechanism underlying increased efficacy. Taken together, we demonstrate the groundbreaking efficacy of capmatinib and radiation as a highly promising concept for future clinical trials.
Collapse
Affiliation(s)
- Marc Zuckermann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg, Germany.
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany.
| | - Chen He
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jared Andrews
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Aditi Bagchi
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Roketa Sloan-Henry
- Center for Pediatric Neurological Disease Research, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Brandon Bianski
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jia Xie
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yingzhe Wang
- Preclinical Pharmacokinetics Shared Resource, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Nathaniel Twarog
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, Departments of BiostatisticsSt. Jude Children's Research Hospital, Memphis, 262 Danny Thomas Place, TN, 38105, USA
| | - Kati J Ernst
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yong Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Xiaoyan Zhu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jennifer K Ocasio
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kaitlin M Budd
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - James Dalton
- Department of Pathology, Departments of PathologySt. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Xiaoyu Li
- Department of Pathology, Departments of PathologySt. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Divyabharathi Chepyala
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Junyuan Zhang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ke Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Laura Hover
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jordan T Roach
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kenneth Chun-Ho Chan
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Nina Hofmann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Peter J McKinnon
- Center for Pediatric Neurological Disease Research, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Burgess B Freeman
- Preclinical Pharmacokinetics Shared Resource, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jason Chiang
- Department of Pathology, Departments of PathologySt. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
- Center Of Excellence in Neuro-Oncology Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
5
|
Prokoph N, Matthews JD, Trigg RM, Montes‐Mojarro IA, Burke GAA, Fend F, Merkel O, Kenner L, Geoerger B, Johnston R, Murray MJ, Riguad C, Brugières L, Turner SD. Patient-derived xenograft models of ALK+ ALCL reveal preclinical promise for therapy with brigatinib. Br J Haematol 2023; 202:985-994. [PMID: 37357529 PMCID: PMC10952693 DOI: 10.1111/bjh.18953] [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: 04/20/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/27/2023]
Abstract
Anaplastic large-cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by the oncogenic anaplastic lymphoma kinase (ALK), accounting for approximately 15% of all paediatric non-Hodgkin lymphoma. Patients with central nervous system (CNS) relapse are particularly difficult to treat with a 3-year overall survival of 49% and a median survival of 23.5 months. The second-generation ALK inhibitor brigatinib shows superior penetration of the blood-brain barrier unlike the first-generation drug crizotinib and has shown promising results in ALK+ non-small-cell lung cancer. However, the benefits of brigatinib in treating aggressive paediatric ALK+ ALCL are largely unknown. We established a patient-derived xenograft (PDX) resource from ALK+ ALCL patients at or before CNS relapse serving as models to facilitate the development of future therapies. We show in vivo that brigatinib is effective in inducing the remission of PDX models of crizotinib-resistant (ALK C1156Y, TP53 loss) ALCL and furthermore that it is superior to crizotinib as a second-line approach to the treatment of a standard chemotherapy relapsed/refractory ALCL PDX pointing to brigatinib as a future therapeutic option.
Collapse
Affiliation(s)
- Nina Prokoph
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
| | - Jamie D. Matthews
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
| | - Ricky M. Trigg
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
| | - Ivonne A. Montes‐Mojarro
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center TübingenUniversity Hospital Tübingen, Eberhard‐Karls‐UniversityTübingenGermany
| | - G. A. Amos Burke
- Department of Paediatric Haematology and OncologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Falko Fend
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center TübingenUniversity Hospital Tübingen, Eberhard‐Karls‐UniversityTübingenGermany
| | - Olaf Merkel
- Department of Experimental Pathology and Laboratory Animal Pathology, Institute of Clinical PathologyMedical University of ViennaViennaAustria
| | - Lukas Kenner
- Department of Experimental Pathology and Laboratory Animal Pathology, Institute of Clinical PathologyMedical University of ViennaViennaAustria
- Unit of Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
- Christian Doppler Laboratory for Applied MetabolomicsMedical University of ViennaViennaAustria
- Center for Biomarker Research in Medicine (CBmed) Vienna, Core‐Lab2Medical University of ViennaViennaAustria
| | - Birgit Geoerger
- Department of Pediatric and Adolescent OncologyGustave Roussy Cancer CenterVillejuifFrance
- INSERM U1015, Gustave Roussy Cancer CenterUniversité Paris‐SaclayVillejuifFrance
| | - Robert Johnston
- Department of Paediatric Oncology/HaematologyRoyal Belfast Hospital for Sick ChildrenBelfastUK
| | - Matthew J. Murray
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
- Department of Paediatric Haematology and OncologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Charlotte Riguad
- Department of Pediatric and Adolescent OncologyGustave Roussy Cancer CenterVillejuifFrance
| | - Laurence Brugières
- Department of Pediatric and Adolescent OncologyGustave Roussy Cancer CenterVillejuifFrance
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of Cambridge, Addenbrooke's HospitalCambridgeUK
- Institute of Medical Genetics and Genomics, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| |
Collapse
|
6
|
Brugières L, Cozic N, Houot R, Rigaud C, Sibon D, Arfi-Rouche J, Bories P, Cottereau AS, Delmer A, Ducassou S, Garnier N, Lamant L, Leruste A, Millot F, Moalla S, Morschhauser F, Nolla M, Pagnier A, Reguerre Y, Renaud L, Schmitt A, Simonin M, Verschuur A, Hoog Labouret N, Mahier Ait Oukhatar C, Vassal G. Efficacy and safety of crizotinib in ALK-positive systemic anaplastic large-cell lymphoma in children, adolescents, and adult patients: results of the French AcSé-crizotinib trial. Eur J Cancer 2023; 191:112984. [PMID: 37549532 DOI: 10.1016/j.ejca.2023.112984] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND The French phase II AcSé-crizotinib trial aimed to evaluate the safety and efficacy of crizotinib in patients with ALK, ROS1, and MET-driven malignancies, including ALK-positive anaplastic large-cell lymphoma (ALK+ ALCL). METHODS ALK+ ALCL patients 12 months or older with measurable disease and no standard care options available received crizotinib twice daily at 165 mg/m2 in children and adolescents and 250 mg in adults. The primary end-point was the response rate at 8 weeks. RESULTS Twenty-eight patients were enroled between February 2014 and March 2018. Three patients who were not treated were excluded from the analysis. The median age was 19 years. The median previous line of chemotherapy was two. In the 24 patients with an evaluable response, the response rate at 8 weeks was 67% (95% CI: 47-82%). All patients discontinued crizotinib after a median treatment duration of 3.7 months: eight for progression, two for adverse events (AEs) related to prior treatments, and 15 by choice, including six for allogeneic stem-cell transplantation. The median follow-up was 45 months. Nine patients experienced an event: eight relapses (seven after crizotinib discontinuation and one after dose reduction), and one died in complete remission. The median duration of response was 43.3 months (95% CI: 8.3-not reached). The 3-year progression-free and overall survival rates were 40% (95% CI: 23-59%) and 63% (95% CI: 43-79%). Grade 3 or 4 treatment-related AEs occurred in 32% of patients. CONCLUSION Crizotinib shows efficacy and an acceptable safety profile in ALK+ ALCL relapsed/refractory patients. However, a large proportion of patients experience a relapse after crizotinib discontinuation. Future studies will assess if prolonged ALK inhibitor exposure has curative potential without consolidation.
Collapse
Affiliation(s)
- Laurence Brugières
- Department of Children and Adolescent Oncology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France.
| | - Nathalie Cozic
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Oncostat U1018 INSERM, Labeled Ligue Contre le Cancer, Université Paris-Saclay, Villejuif, France
| | - Roch Houot
- Department of Hematology, CHU de Rennes, Université de Rennes, Rennes, France
| | - Charlotte Rigaud
- Department of Children and Adolescent Oncology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - David Sibon
- Lymphoid Malignancies Department, Henri Mondor University Hospital, AP-HP, Creteil, France
| | - Julia Arfi-Rouche
- Department of Radiology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Pierre Bories
- Institut Universitaire du Cancer - Oncopole, Toulouse, France
| | - Anne S Cottereau
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, University of Paris, Paris, France
| | - Alain Delmer
- Department of Hematology, University Hospital of Reims and UFR Médecine, Reims, France
| | | | - Nathalie Garnier
- Institut d'Hematologie et d'Oncologie Pediatrique, Hospices Civils de Lyon, Lyon, France
| | - Laurence Lamant
- Department of Pathology, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France Université Toulouse III-Paul Sabatier; UMR1037 CRCT, Toulouse, France
| | - Amaury Leruste
- SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), Institut Curie, PSL Research University, Paris, France
| | | | - S Moalla
- Institut Universitaire du Cancer - Oncopole, Toulouse, France
| | - Franck Morschhauser
- ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, Univ. Lille, CHU Lille, Lille, France
| | - Marie Nolla
- Pediatric Hematology-Immunology, CHU Toulouse Purpan, France
| | - Anne Pagnier
- Pediatric Immunology Hematology and Oncology, CHU Grenoble Alpes, France
| | - Yves Reguerre
- CHU de Saint Denis de La Réunion Service d'Oncologie et d'Hématologie Pédiatrique, Saint Denis, France
| | - Loic Renaud
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-Oncologie, DMU DHI; Université de Paris, Paris, France
| | - Anne Schmitt
- Hématologie, Institut Bergonié, Bordeaux, France
| | - Mathieu Simonin
- Department of Pediatric Hematology and Oncology, Assistance Publique-Hôpitaux de Paris Armand Trousseau Hospital, Sorbonne Université, Paris, France
| | - Arnaud Verschuur
- Department of Pediatric Hematology-Oncology, La Timone University Hospital, APHM, Marseille, France
| | | | | | - Gilles Vassal
- Department of Children and Adolescent Oncology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| |
Collapse
|
7
|
Burikhanov R, Ganguly S, Ellingson S, Sviripa VM, Araujo N, Li S, Venkatraman P, Rao M, Choughule A, Brainson CF, Zhan CG, Spielmann HP, Watt DS, Govindan R, Rangnekar VM. Crizotinib induces Par-4 secretion from normal cells and GRP78 expression on the cancer cell surface for selective tumor growth inhibition. Am J Cancer Res 2023; 13:976-991. [PMID: 37034206 PMCID: PMC10077052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/20/2023] [Indexed: 04/11/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths. Lung cancer cells develop resistance to apoptosis by suppressing the secretion of the tumor suppressor Par-4 protein (also known as PAWR) and/or down-modulating the Par-4 receptor GRP78 on the cell surface (csGRP78). We sought to identify FDA-approved drugs that elevate csGRP78 on the surface of lung cancer cells and induce Par-4 secretion from the cancer cells and/or normal cells in order to inhibit cancer growth in an autocrine or paracrine manner. In an unbiased screen, we identified crizotinib (CZT), an inhibitor of activated ALK/MET/ROS1 receptor tyrosine kinase, as an inducer of csGRP78 expression in ALK-negative, KRAS or EGFR mutant lung cancer cells. Elevation of csGRP78 in the lung cancer cells was dependent on activation of the non-receptor tyrosine kinase SRC by CZT. Inhibition of SRC activation in the cancer cells prevented csGRP78 translocation but promoted Par-4 secretion by CZT, implying that activated SRC prevented Par-4 secretion. In normal cells, CZT did not activate SRC and csGRP78 elevation but induced Par-4 secretion. Consequently, CZT induced Par-4 secretion from normal cells and elevated csGRP78 in the ALK-negative tumor cells to cause paracrine apoptosis in cancer cell cultures and growth inhibition of tumor xenografts in mice. Thus, CZT induces differential activation of SRC in normal and cancer cells to trigger the pro-apoptotic Par-4-GRP78 axis. As csGRP78 is a targetable receptor, CZT can be repurposed to elevate csGRP78 for inhibition of ALK-negative lung tumors.
Collapse
Affiliation(s)
- Ravshan Burikhanov
- Department of Radiation Medicine, College of Medicine, University of KentuckyLexington, Kentucky, USA
| | - Saptadwipa Ganguly
- Department of Toxicology and Cancer Biology, College of Medicine, University of KentuckyLexington, Kentucky, USA
| | - Sally Ellingson
- Department of Internal Medicine, College of Medicine, University of KentuckyLexington, Kentucky, USA
| | - Vitaliy M Sviripa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of KentuckyLexington, Kentucky, USA
| | - Nathalia Araujo
- Department of Toxicology and Cancer Biology, College of Medicine, University of KentuckyLexington, Kentucky, USA
| | - Shunqiang Li
- Department of Medicine, Division of Oncology, Washington UniversitySt. Louis, Missouri, USA
| | - Prasanna Venkatraman
- Tata Memorial Centre-Advanced Centre for Treatment Research and Education in CancerNavi Mumbai, Maharashtra, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher EducationManipal, Karnataka, India
| | - Anuradha Choughule
- Tata Memorial Centre-Advanced Centre for Treatment Research and Education in CancerNavi Mumbai, Maharashtra, India
| | - Christine F Brainson
- Department of Toxicology and Cancer Biology, College of Medicine, University of KentuckyLexington, Kentucky, USA
- Markey Cancer Center, University of KentuckyLexington, Kentucky, USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of KentuckyLexington, Kentucky, USA
| | - H Peter Spielmann
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of KentuckyLexington, Kentucky, USA
| | - David S Watt
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of KentuckyLexington, Kentucky, USA
| | - Ramaswamy Govindan
- Department of Medicine, Division of Oncology, Washington UniversitySt. Louis, Missouri, USA
| | - Vivek M Rangnekar
- Department of Radiation Medicine, College of Medicine, University of KentuckyLexington, Kentucky, USA
- Markey Cancer Center, University of KentuckyLexington, Kentucky, USA
| |
Collapse
|
8
|
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
|
9
|
Merino M, Kasamon Y, Li H, Ma L, Leong R, Zhou J, Reaman G, Chambers W, Richardson N, Theoret M, Pazdur R, Gormley N. FDA approval summary: Crizotinib for pediatric and young adult patients with relapsed or refractory systemic anaplastic large cell lymphoma. Pediatr Blood Cancer 2022; 69:e29602. [PMID: 35561013 DOI: 10.1002/pbc.29602] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/07/2022] [Accepted: 01/27/2022] [Indexed: 12/14/2022]
Abstract
In January 2021, the U.S. Food and Drug Administration (FDA) approved crizotinib for pediatric patients 1 year and older and young adults with relapsed or refractory systemic anaplastic large cell lymphoma (sALCL). This is the first approval for pediatric sALCL. Approval was based on a single-arm trial of crizotinib monotherapy that included 26 patients, aged 1-20 years, with previously treated sALCL. Efficacy was based on centrally assessed objective response rate (88%) and duration of response. Herein, we highlight unique aspects of the regulatory review, including extension of the indication to young adults, postmarketing safety, and dose optimization strategies.
Collapse
Affiliation(s)
- Margret Merino
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yvette Kasamon
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hongshan Li
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lian Ma
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ruby Leong
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiaxi Zhou
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gregory Reaman
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA.,Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Wiley Chambers
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nicholas Richardson
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Marc Theoret
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA.,Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Richard Pazdur
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA.,Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nicole Gormley
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
10
|
Parrish RH, Ashworth LD, Löbenberg R, Benavides S, Cies JJ, MacArthur RB. Compounded Nonsterile Preparations and FDA-Approved Commercially Available Liquid Products for Children: A North American Update. Pharmaceutics 2022; 14:1032. [PMID: 35631618 PMCID: PMC9144535 DOI: 10.3390/pharmaceutics14051032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 11/16/2022] Open
Abstract
The purpose of this work was to evaluate the suitability of recent US Food and Drug Administration (US-FDA)-approved and marketed oral liquid, powder, or granule products for children in North America, to identify the next group of Active Pharmaceutical Ingredients (APIs) that have high potential for development as commercially available FDA-approved finished liquid dosage forms, and to propose lists of compounded nonsterile preparations (CNSPs) that should be developed as commercially available FDA-approved finished liquid dosage forms, as well as those that pharmacists should continue to compound extemporaneously. Through this identification and categorization process, the pharmaceutical industry, government, and professionals are encouraged to continue to work together to improve the likelihood that patients will receive high-quality standardized extemporaneously compounded CNSPs and US-FDA-approved products.
Collapse
Affiliation(s)
- Richard H. Parrish
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA 31902, USA
| | - Lisa D. Ashworth
- Department of Pharmacy Services, Children’s Health System of Texas, Dallas, TX 75235, USA;
| | - Raimar Löbenberg
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Sandra Benavides
- School of Pharmacy, Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, USA;
| | - Jeffrey J. Cies
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA 19129, USA;
- Department of Pharmacy Services, St. Christopher’s Hospital for Children/Tower Health, Philadelphia, PA 19134, USA
| | - Robert B. MacArthur
- Department of Pharmacy Services, Rockefeller University Hospital, New York, NY 10065, USA;
| |
Collapse
|
11
|
Yang F, Wu H, Bo Y, Lu Y, Pan H, Li S, Lu Q, Xie S, Liao H, Wang B. Population Pharmacokinetic Modeling and Simulation of TQ-B3101 to Inform Dosing in Pediatric Patients With Solid Tumors. Front Pharmacol 2022; 12:782518. [PMID: 35115931 PMCID: PMC8804354 DOI: 10.3389/fphar.2021.782518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/14/2021] [Indexed: 11/18/2022] Open
Abstract
Background: TQ-B3101 is a novel kinase inhibitor currently in development for the treatment of advanced malignant solid tumor and relapsed or refractory ALK-positive anaplastic large cell lymphoma. Methods: A population pharmacokinetic model was developed using data collected from a Phase 1 study and a Phase 2 study to characterize the pharmacokinetic of TQ-B3101 and its active metabolite (TQ-B3101M). The final model was used to optimize dosing of TQ-B3101 for pediatric patients (6-<18 years) with anaplastic large cell lymphoma. Results: The pharmacokinetic of TQ-B3101 and TQ-B3101M was adequately described by a 1-compartment model with first-order absorption and elimination for parent drug coupled with a 2-compartment model with time-dependent clearance for the metabolite. The clearance of TQ-B3101M decreased over time with a maximum fractional reduction of 0.41. The estimated apparent clearance and apparent volume of distribution of TQ-B3101 were 2850 L/h and 4200 L, respectively. The elimination half-life of TQ-B3101 was 1.0 h. The distribution and elimination half-lives of TQ-B3101M at steady state were 4.9 and 39.4 h, respectively. The projected exposure of TQ-B3101M in virtual pediatric population following the body surface area tiered dosing regimen was similar to that in children pediatric patients after the recommended pediatric dose of crizotinib (280 mg/m2 twice daily), an analog of TQ-B3101M. Conclusion: A population pharmacokinetic model was developed to provide optimal dose of regimen for further development of TQ-B3101 in pediatric patients with anaplastic large cell lymphoma.
Collapse
Affiliation(s)
- Fen Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing, China
- *Correspondence: Fen Yang,
| | - Huali Wu
- Amador Bioscience, Hangzhou, China
| | - Yunhai Bo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ye Lu
- Amador Bioscience, Hangzhou, China
| | - Hong Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Su Li
- Department of Clinical Trial Center, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Qin Lu
- Chia Tai Tianqing Pharmaceutical Group CO., Ltd., Nanjing, China
| | | | | | | |
Collapse
|
12
|
Grundy M, Narendran A. The hepatocyte growth factor/mesenchymal epithelial transition factor axis in high-risk pediatric solid tumors and the anti-tumor activity of targeted therapeutic agents. Front Pediatr 2022; 10:910268. [PMID: 36034555 PMCID: PMC9399617 DOI: 10.3389/fped.2022.910268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/15/2022] [Indexed: 01/04/2023] Open
Abstract
Clinical trials completed in the last two decades have contributed significantly to the improved overall survival of children with cancer. In spite of these advancements, disease relapse still remains a significant cause of death in this patient population. Often, increasing the intensity of current protocols is not feasible because of cumulative toxicity and development of drug resistance. Therefore, the identification and clinical validation of novel targets in high-risk and refractory childhood malignancies are essential to develop effective new generation treatment protocols. A number of recent studies have shown that the hepatocyte growth factor (HGF) and its receptor Mesenchymal epithelial transition factor (c-MET) influence the growth, survival, angiogenesis, and metastasis of cancer cells. Therefore, the c-MET receptor tyrosine kinase and HGF have been identified as potential targets for cancer therapeutics and recent years have seen a race to synthesize molecules to block their expression and function. In this review we aim to summarize the literature that explores the potential and biological rationale for targeting the HGF/c-MET pathway in common and high-risk pediatric solid tumors. We also discuss selected recent and ongoing clinical trials with these agents in relapsed pediatric tumors that may provide applicable future treatments for these patients.
Collapse
Affiliation(s)
- Megan Grundy
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aru Narendran
- POETIC Laboratory for Preclinical and Drug Discovery Studies, Division of Pediatric Oncology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
13
|
Resistance to Targeted Agents Used to Treat Paediatric ALK-Positive ALCL. Cancers (Basel) 2021; 13:cancers13236003. [PMID: 34885113 PMCID: PMC8656581 DOI: 10.3390/cancers13236003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary In general, the non-Hodgkin lymphoma (NHL), anaplastic large cell lymphoma (ALCL) diagnosed in childhood has a good survival outcome when treated with multi-agent chemotherapy. However, side effects of treatment are common, and outcomes are poorer after relapse, which occurs in up to 30% of cases. New drugs are required that are more effective and have fewer side effects. Targeted therapies are potential solutions to these problems, however, the development of resistance may limit their impact. This review summarises the potential resistance mechanisms to these targeted therapies. Abstract Non-Hodgkin lymphoma (NHL) is the third most common malignancy diagnosed in children. The vast majority of paediatric NHL are either Burkitt lymphoma (BL), diffuse large B-cell lymphoma (DLBCL), anaplastic large cell lymphoma (ALCL), or lymphoblastic lymphoma (LL). Multi-agent chemotherapy is used to treat all of these types of NHL, and survival is over 90% but the chemotherapy regimens are intensive, and outcomes are generally poor if relapse occurs. Therefore, targeted therapies are of interest as potential solutions to these problems. However, the major problem with all targeted agents is the development of resistance. Mechanisms of resistance are not well understood, but increased knowledge will facilitate optimal management strategies through improving our understanding of when to select each targeted agent, and when a combinatorial approach may be helpful. This review summarises currently available knowledge regarding resistance to targeted therapies used in paediatric anaplastic lymphoma kinase (ALK)-positive ALCL. Specifically, we outline where gaps in knowledge exist, and further investigation is required in order to find a solution to the clinical problem of drug resistance in ALCL.
Collapse
|
14
|
Gibson EG, Campagne O, Selvo NS, Gajjar A, Stewart CF. Population pharmacokinetic analysis of crizotinib in children with progressive/recurrent high-grade and diffuse intrinsic pontine gliomas. Cancer Chemother Pharmacol 2021; 88:1009-1020. [PMID: 34586478 DOI: 10.1007/s00280-021-04357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/19/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Crizotinib, a potent oral tyrosine kinase inhibitor, was evaluated in combination with dasatinib in a phase 1 trial (NCT01644773) in children with progressive or recurrent high-grade and diffuse intrinsic pontine gliomas (HGG and DIPG). This study aimed to characterize the pharmacokinetics of crizotinib in this population and identify significant covariates. METHODS Patients (N = 36, age range 2.9-21.3 years) were treated orally once or twice-daily with 100-215 mg/m2 crizotinib and 50-65 mg/m2 dasatinib. Pharmacokinetic studies were performed for crizotinib alone after the first dose and at steady state, and for the drug combination at steady state. Crizotinib plasma concentrations were measured using a validated LC-MS/MS method. Population modeling was performed (Monolix) and the impact of factors including patient demographics and co-medications were investigated on crizotinib pharmacokinetics. RESULTS Crizotinib concentrations were described with a linear two-compartment model and absorption lag time. Concomitant dasatinib and overweight/obese status significantly influenced crizotinib pharmacokinetics, resulting in clinically relevant impact (> 20%) on drug exposure. Crizotinib mean apparent clearance (CL/F) was 66.7 L/h/m2 after single-dose and decreased to 26.5 L/h/m2 at steady state when given alone, but not when combined with dasatinib (mean 60.8 L/h/m2). Overweight/obese patients exhibited lower crizotinib CL/F and apparent volume V1/F (mean 46.2 L/h/m2 and 73.3 L/m2) compared to other patients (mean 75.5 L/h/m2 and 119.3 L/m2, p < 0.001). CONCLUSION A potential pharmacokinetic interaction was observed between crizotinib and dasatinib in children with HGG and DIPG. Further, crizotinib exposure was significantly higher in overweight/obese patients, who may require a dosing adjustment.
Collapse
Affiliation(s)
- Elizabeth G Gibson
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.,Bristol Myers Squibb, Princeton, NJ, USA
| | - Olivia Campagne
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Nicholas S Selvo
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Amar Gajjar
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Clinton F Stewart
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
15
|
IL10RA modulates crizotinib sensitivity in NPM1-ALK+ anaplastic large cell lymphoma. Blood 2021; 136:1657-1669. [PMID: 32573700 DOI: 10.1182/blood.2019003793] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/19/2020] [Indexed: 02/08/2023] Open
Abstract
Anaplastic large cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by a hyperactive anaplastic lymphoma kinase (ALK) fusion protein. ALK inhibitors, such as crizotinib, provide alternatives to standard chemotherapy with reduced toxicity and side effects. Children with lymphomas driven by nucleophosmin 1 (NPM1)-ALK fusion proteins achieved an objective response rate to ALK inhibition therapy of 54% to 90% in clinical trials; however, a subset of patients progressed within the first 3 months of treatment. The mechanism for the development of ALK inhibitor resistance is unknown. Through genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) activation and knockout screens in ALCL cell lines, combined with RNA sequencing data derived from ALK inhibitor-relapsed patient tumors, we show that resistance to ALK inhibition by crizotinib in ALCL can be driven by aberrant upregulation of interleukin 10 receptor subunit alpha (IL10RA). Elevated IL10RA expression rewires the STAT3 signaling pathway, bypassing otherwise critical phosphorylation by NPM1-ALK. IL-10RA expression does not correlate with response to standard chemotherapy in pediatric patients, suggesting that a combination of crizotinib and chemotherapy could prevent ALK inhibitor resistance-specific relapse.
Collapse
|
16
|
Foster JH, Voss SD, Hall DC, Minard CG, Balis FM, Wilner K, Berg SL, Fox E, Adamson PC, Blaney SM, Weigel BJ, Mossé YP. Activity of Crizotinib in Patients with ALK-Aberrant Relapsed/Refractory Neuroblastoma: A Children's Oncology Group Study (ADVL0912). Clin Cancer Res 2021; 27:3543-3548. [PMID: 33568345 DOI: 10.1158/1078-0432.ccr-20-4224] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 02/04/2021] [Indexed: 12/27/2022]
Abstract
PURPOSE Anaplastic lymphoma kinase (ALK) aberrations are a promising target for patients with neuroblastoma. We assessed the activity of first-generation ALK inhibitor crizotinib in patients with no known curative treatments and whose tumors harbored an activating ALK alteration. PATIENTS AND METHODS Twenty patients with relapsed/refractory ALK-positive neuroblastoma received crizotinib at the recommended phase II dose of 280 mg/m2/dose. A Simon two-stage design was used to evaluate the antitumor activity of crizotinib monotherapy. Response evaluation occurred after cycles 1, 3, 5, 7, and then every 3 cycles. Correlation of ALK status and response was a secondary aim of the study. RESULTS The objective response rate for patients with neuroblastoma was 15% [95% confidence interval (CI): 3.3%-34.3%]: two with partial responses and 1 with a complete response. All three patients had a somatic ALK Arg1275Gln mutation, the most common ALK hotspot mutation observed in neuroblastoma and the only mutation predicted to be sensitive to ALK inhibition with crizotinib. Two patients had prolonged stable disease (10 and 13 cycles, respectively); both harbored an ALK Arg1275Gln mutation. Three patients with ALK Phe1174Leu mutations progressed during cycle 1 of therapy, and one patient with an ALK Phe1174Val received three cycles before disease progression. The two patients with ALK amplification had no response. The most common adverse event was a decrease in neutrophil count. CONCLUSIONS Despite limited activity seen in this trial, we conclude that this is more likely due to an inability to reach the higher concentrations of crizotinib needed to overcome the competing ATP affinity.See related commentary by Schulte and Eggert, p. 3507.
Collapse
Affiliation(s)
- Jennifer H Foster
- Baylor College of Medicine; Texas Children's Cancer and Hematology Centers, Houston, Texas
| | - Stephan D Voss
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | | | - Charles G Minard
- Baylor College of Medicine; Texas Children's Cancer and Hematology Centers, Houston, Texas
| | - Frank M Balis
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Stacey L Berg
- Baylor College of Medicine; Texas Children's Cancer and Hematology Centers, Houston, Texas
| | - Elizabeth Fox
- St Jude Children's Research Hospital, Memphis, Tennessee
| | - Peter C Adamson
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan M Blaney
- Baylor College of Medicine; Texas Children's Cancer and Hematology Centers, Houston, Texas
| | | | - Yael P Mossé
- Division of Oncology and Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. .,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Greengard E, Mosse YP, Liu X, Minard CG, Reid JM, Voss S, Wilner K, Fox E, Balis F, Blaney SM, Adamson PC, Weigel BJ. Safety, tolerability and pharmacokinetics of crizotinib in combination with cytotoxic chemotherapy for pediatric patients with refractory solid tumors or anaplastic large cell lymphoma (ALCL): a Children's Oncology Group phase 1 consortium study (ADVL1212). Cancer Chemother Pharmacol 2020; 86:829-840. [PMID: 33095287 DOI: 10.1007/s00280-020-04171-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/06/2020] [Indexed: 02/10/2023]
Abstract
PURPOSE This phase 1 study aimed to determine the safety, tolerability and recommended phase 2 dose (RP2D) of crizotinib in combination with cytotoxic chemotherapy for children with refractory solid tumors and ALCL. METHODS Pediatric patients with treatment refractory solid tumors or ALCL were eligible. Using a 3 + 3 design, crizotinib was escalated in three dose levels: 165, 215, or 280 mg/m2/dose BID. In Part A, patients received crizotinib oral solution (OS) in combination with topotecan and cyclophosphamide (topo/cyclo); in Part B, crizotinib OS was administered with vincristine and doxorubicin (vcr/dox). In Parts C and D, patients received topo/cyclo in combination with either crizotinib-formulated capsules (FC) or microspheres (cMS), respectively. Crizotinib pharmacokinetic evaluation was required. RESULTS Forty-four eligible patients were enrolled, 39 were evaluable for toxicity. Parts A and B were terminated due to concerns regarding palatability and tolerability of the OS. In Part C, crizotinib, FC 215 mg/m2/dose BID, in combination with topo/cyclo was tolerated. In Part D, the maximum tolerated dose (MTD) was exceeded at 165 mg/m2/dose of crizotinib cMS. Pharmacokinetics of crizotinib in combination with chemotherapy was similar to single-agent crizotinib and exposures were not formulation dependent. CONCLUSIONS The RP2D of crizotinib FCs in combination with cyclophosphamide and topotecan was 215 mg/m2/dose BID. The oral solution of crizotinib was not palatable in this patient population. Crizotinib cMS was palatable; however, patients experienced increased toxicity that was not explained by the relative bioavailability or exposure and warrants further investigation. CLINICAL TRIAL REGISTRY The trial is registered as NCT01606878 at Clinicaltrials.gov.
Collapse
Affiliation(s)
- Emily Greengard
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| | - Yael P Mosse
- St Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaowei Liu
- Children's Oncology Group, Monrovia, CA, USA
| | | | | | | | | | - Elizabeth Fox
- St Jude Children's Research Hospital, Memphis, TN, USA
| | - Frank Balis
- St Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | - Brenda J Weigel
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
18
|
López Quiñones AJ, Wagner DJ, Wang J. Characterization of Meta-Iodobenzylguanidine (mIBG) Transport by Polyspecific Organic Cation Transporters: Implication for mIBG Therapy. Mol Pharmacol 2020; 98:109-119. [PMID: 32487736 PMCID: PMC7330676 DOI: 10.1124/mol.120.119495] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Radiolabeled meta-iodobenzylguanidine (mIBG) is an important radiopharmaceutical used in the diagnosis and treatment of neuroendocrine cancers. mIBG is known to enter tumor cells through the norepinephrine transporter. Whole-body scintigraphy has shown rapid mIBG elimination through the kidney and high accumulation in several normal tissues, but the underlying molecular mechanisms are unclear. Using transporter-expressing cell lines, we show that mIBG is an excellent substrate for human organic cation transporters 1-3 (hOCT1-3) and the multidrug and toxin extrusion proteins 1 and 2-K (hMATE1/2-K), but not for the renal organic anion transporter 1 and 3 (hOAT1/3). Kinetic analysis revealed that hOCT1, hOCT2, hOCT3, hMATE1, and hMATE2-K transport mIBG with similar apparent affinities (K m of 19.5 ± 6.9, 17.2 ± 2.8, 14.5 ± 7.1, 17.7 ± 10.9, 12.6 ± 5.6 µM, respectively). Transwell studies in hOCT2/hMATE1 double-transfected Madin-Darby canine kidney cells showed that mIBG transport in the basal (B)-to-apical (A) direction is much greater than in the A-to-B direction. Compared with control cells, the B-to-A permeability of mIBG increased by 20-fold in hOCT2/hMATE1 double-transfected cells. Screening of 23 drugs used in the treatment of neuroblastoma identified several drugs with the potential to inhibit hOCT- or hMATE-mediated mIBG uptake. Interestingly, irinotecan selectively inhibited hOCT1, whereas crizotinib potently inhibited hOCT3-mediated mIBG uptake. Our results suggest that mIBG undergoes renal tubular secretion mediated by hOCT2 and hMATE1/2-K, and hOCT1 and hOCT3 may play important roles in mIBG uptake into normal tissues. SIGNIFICANCE STATEMENT: mIBG is eliminated by the kidney and extensively accumulates in several tissues known to express hOCT1 and hOCT3. Our results suggest that hOCT2 and human multidrug and toxin extrusion proteins 1 and 2-K are involved in mIBG renal elimination, whereas hOCT1 and hOCT3 may play important roles in mIBG uptake into normal tissues. These findings may help to predict and prevent adverse drug interaction with therapeutic [131I]mIBG and develop clinical strategies to reduce [131I]mIBG accumulation and toxicity in normal tissues and organs.
Collapse
Affiliation(s)
| | - David J Wagner
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
19
|
Janssen JM, Dorlo TPC, Steeghs N, Beijnen JH, Hanff LM, van Eijkelenburg NKA, van der Lugt J, Zwaan CM, Huitema ADR. Pharmacokinetic Targets for Therapeutic Drug Monitoring of Small Molecule Kinase Inhibitors in Pediatric Oncology. Clin Pharmacol Ther 2020; 108:494-505. [PMID: 32022898 DOI: 10.1002/cpt.1808] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
In recent years new targeted small molecule kinase inhibitors have become available for pediatric patients with cancer. Relationships between drug exposure and treatment response have been established for several of these drugs in adults. Following these exposure-response relationships, pharmacokinetic (PK) target minimum plasma rug concentration at the end of a dosing interval (Cmin ) values to guide therapeutic drug monitoring (TDM) in adults have been proposed. Despite the fact that variability in PK may be even larger in pediatric patients, TDM is only sparsely applied in pediatric oncology. Based on knowledge of the PK, mechanism of action, molecular driver, and pathophysiology of the disease, we bridge available data on the exposure-efficacy relationship from adults to children and propose target Cmin values to guide TDM for the pediatric population. Dose adjustments in individual pediatric patients can be based on these targets. Nevertheless, further research should be performed to validate these targets.
Collapse
Affiliation(s)
- Julie M Janssen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thomas P C Dorlo
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Clinical Pharmacology, Division of Medical Oncology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, Amsterdam, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Lidwien M Hanff
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | | | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Hematology and Oncology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
20
|
Trahair T, Gifford AJ, Fordham A, Mayoh C, Fadia M, Lukeis R, Wood AC, Valvi S, Walker RD, Blackburn J, Heyer EE, Mercer TR, Barbaric D, Marshall GM, MacKenzie KL. Crizotinib and Surgery for Long-Term Disease Control in Children and Adolescents With ALK-Positive Inflammatory Myofibroblastic Tumors. JCO Precis Oncol 2019; 3:1800297. [PMID: 32914017 PMCID: PMC7446396 DOI: 10.1200/po.18.00297] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Before anaplastic lymphoma kinase (ALK) inhibitors, treatment options for ALK-positive inflammatory myofibroblastic tumors (AP-IMTs) were unsatisfactory. We retrospectively analyzed the outcome of patients with AP-IMT treated with crizotinib to document response, toxicity, survival, and features associated with relapse. METHODS The cohort comprised eight patients with AP-IMT treated with crizotinib and surgery. Outcome measures were progression-free and overall survival after commencing crizotinib, treatment-related toxicities, features associated with relapse, outcome after relapse, and outcome after ceasing crizotinib. RESULTS The median follow-up after commencing crizotinib was 3 years (range, 0.9 to 5.5 years). The major toxicity was neutropenia. All patients responded to crizotinib. Five were able to discontinue therapy without recurrence (median treatment duration, 1 year; range, 0.2 to 3.0 years); one continues on crizotinib. Two critically ill patients with initial complete response experienced relapse while on therapy. Both harbored RANBP2-ALK fusions and responded to alternative ALK inhibitors; one ultimately died as a result of progressive disease, whereas the other remains alive on treatment. Progression-free and overall survival since commencement of crizotinib is 0.75 ± 0.15% and 0.83 ± 0.15%, respectively. CONCLUSION We confirm acceptable toxicity and excellent disease control in patients with AP-IMT treated with crizotinib, which may be ceased without recurrence in most. Relapses occurred in two of three patients with RANBP2-ALK translocated IMT, which suggests that such patients require additional therapy.
Collapse
Affiliation(s)
- Toby Trahair
- Sydney Children's Hospital, Randwick, New South Wales, Australia.,Children's Cancer Institute, Sydney, New South Wales, Australia.,University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Sydney, New South Wales, Australia.,Prince of Wales Hospital, Randwick, New South Wales, Australia
| | | | - Chelsea Mayoh
- Children's Cancer Institute, Sydney, New South Wales, Australia
| | - Mitali Fadia
- Canberra Hospital, Garran, Australian Capital Territory, Australia.,Australian National University Medical School, Acton, Australian Capital Territory, Australia
| | - Robyn Lukeis
- St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | | | - Santosh Valvi
- Perth Children's Hospital, Perth, Western Australia, Australia
| | - Roderick D Walker
- Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - James Blackburn
- University of New South Wales, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Erin E Heyer
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Tim R Mercer
- University of New South Wales, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,Altius Institute for Biomedical Sciences, Seattle, WA
| | - Draga Barbaric
- Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Glenn M Marshall
- Sydney Children's Hospital, Randwick, New South Wales, Australia.,Children's Cancer Institute, Sydney, New South Wales, Australia.,University of New South Wales, Sydney, New South Wales, Australia
| | - Karen L MacKenzie
- Children's Medical Research Institute, Westmead New South Wales, Australia
| |
Collapse
|
21
|
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: 40] [Impact Index Per Article: 5.7] [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
|
22
|
Nakagawara A, Li Y, Izumi H, Muramori K, Inada H, Nishi M. Neuroblastoma. Jpn J Clin Oncol 2018; 48:214-241. [PMID: 29378002 DOI: 10.1093/jjco/hyx176] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common solid tumors in children and has a diverse clinical behavior that largely depends on the tumor biology. Neuroblastoma exhibits unique features, such as early age of onset, high frequency of metastatic disease at diagnosis in patients over 1 year of age and the tendency for spontaneous regression of tumors in infants. The high-risk tumors frequently have amplification of the MYCN oncogene as well as segmental chromosome alterations with poor survival. Recent advanced genomic sequencing technology has revealed that mutation of ALK, which is present in ~10% of primary tumors, often causes familial neuroblastoma with germline mutation. However, the frequency of gene mutations is relatively small and other aberrations, such as epigenetic abnormalities, have also been proposed. The risk-stratified therapy was introduced by the Japan Neuroblastoma Study Group (JNBSG), which is now moving to the Neuroblastoma Committee of Japan Children's Cancer Group (JCCG). Several clinical studies have facilitated the reduction of therapy for children with low-risk neuroblastoma disease and the significant improvement of cure rates for patients with intermediate-risk as well as high-risk disease. Therapy for patients with high-risk disease includes intensive induction chemotherapy and myeloablative chemotherapy, followed by the treatment of minimal residual disease using differentiation therapy and immunotherapy. The JCCG aims for better cures and long-term quality of life for children with cancer by facilitating new approaches targeting novel driver proteins, genetic pathways and the tumor microenvironment.
Collapse
Affiliation(s)
| | - Yuanyuan Li
- Laboratory of Molecular Biology, Life Science Research Institute, Saga Medical Center Koseikan
| | - Hideki Izumi
- Laboratory of Molecular Biology, Life Science Research Institute, Saga Medical Center Koseikan
| | | | - Hiroko Inada
- Department of Pediatrics, Saga Medical Center Koseikan
| | - Masanori Nishi
- Department of Pediatrics, Saga University, Saga 849-8501, Japan
| |
Collapse
|
23
|
ALK in Neuroblastoma: Biological and Therapeutic Implications. Cancers (Basel) 2018; 10:cancers10040113. [PMID: 29642598 PMCID: PMC5923368 DOI: 10.3390/cancers10040113] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 01/09/2023] Open
Abstract
Neuroblastoma (NB) is the most common and deadly solid tumour in children. Despite the development of new treatment options for high-risk NB, over half of patients relapse and five-year survival remains at 40-50%. Therefore, novel treatment strategies aimed at providing long-term disease remission are urgently sought. ALK, encoding the anaplastic lymphoma kinase receptor, is altered by gain-of-function point mutations in around 14% of high-risk NB and represents an ideal therapeutic target given its low or absent expression in healthy tissue postnatally. Small-molecule inhibitors of Anaplastic Lymphoma Kinase (ALK) approved in ALK fusion-positive lung cancer are currently undergoing clinical assessment in patients with ALK-mutant NB. Parallel pre-clinical studies are demonstrating the efficacy of ALK inhibitors against common ALK variants in NB; however, a complex picture of therapeutic resistance is emerging. It is anticipated that long-term use of these compounds will require combinatorial targeting of pathways downstream of ALK, functionally-related 'bypass' mechanisms and concomitant oncogenic pathways.
Collapse
|
24
|
Theilen TM, Soerensen J, Bochennek K, Becker M, Schwabe D, Rolle U, Klingebiel T, Lehrnbecher T. Crizotinib in ALK + inflammatory myofibroblastic tumors-Current experience and future perspectives. Pediatr Blood Cancer 2018; 65. [PMID: 29286567 DOI: 10.1002/pbc.26920] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/28/2017] [Accepted: 11/08/2017] [Indexed: 02/02/2023]
Abstract
Inflammatory myofibroblastic tumor (IMT) and its subtype epithelioid inflammatory myofibroblastic sarcoma (EIMS) are rare soft-tissue tumors. As about 50% of IMT and 100% of EIMS contain activating rearrangements of the anaplastic lymphoma kinase (ALK) gene, targeted kinase inhibition of ALK by compounds such as crizotinib is a potential treatment option. We performed a literature review and analyzed a total of 30 patients with IMT/EIMS treated with crizotinib. A total of 12 patients achieved complete or partial remission. As preliminary data are promising, a prospective study evaluating crizotinib treatment in patients with unresectable/multifocal ALK+ IMT/EIMS is warranted.
Collapse
Affiliation(s)
- Till-Martin Theilen
- Department of Pediatric Surgery and Pediatric Urology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Jan Soerensen
- Division of Pediatric Stem Cell Transplantation and Immunology, Hospital for Children and Adolescents*, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Konrad Bochennek
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Martina Becker
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Dirk Schwabe
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery and Pediatric Urology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Thomas Klingebiel
- Division of Pediatric Stem Cell Transplantation and Immunology, Hospital for Children and Adolescents*, Johann Wolfgang Goethe University, Frankfurt, Germany
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Thomas Lehrnbecher
- Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
25
|
Treatment Options for Paediatric Anaplastic Large Cell Lymphoma (ALCL): Current Standard and beyond. Cancers (Basel) 2018; 10:cancers10040099. [PMID: 29601554 PMCID: PMC5923354 DOI: 10.3390/cancers10040099] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 01/22/2023] Open
Abstract
Anaplastic Lymphoma Kinase (ALK)-positive Anaplastic Large Cell Lymphoma (ALCL), remains one of the most curable cancers in the paediatric setting; multi-agent chemotherapy cures approximately 65–90% of patients. Over the last two decades, major efforts have focused on improving the survival rate by intensification of combination chemotherapy regimens and employing stem cell transplantation for chemotherapy-resistant patients. More recently, several new and ‘renewed’ agents have offered the opportunity for a change in the paradigm for the management of both chemo-sensitive and chemo-resistant forms of ALCL. The development of ALK inhibitors following the identification of the EML4-ALK fusion gene in Non-Small Cell Lung Cancer (NSCLC) has opened new possibilities for ALK-positive ALCL. The uniform expression of CD30 on the cell surface of ALCL has given the opportunity for anti-CD30 antibody therapy. The re-evaluation of vinblastine, which has shown remarkable activity as a single agent even in the face of relapsed disease, has led to the consideration of a revised approach to frontline therapy. The advent of immune therapies such as checkpoint inhibition has provided another option for the treatment of ALCL. In fact, the number of potential new agents now presents a real challenge to the clinical community that must prioritise those thought to offer the most promise for the future. In this review, we will focus on the current status of paediatric ALCL therapy, explore how new and ‘renewed’ agents are re-shaping the therapeutic landscape for ALCL, and identify the strategies being employed in the next generation of clinical trials.
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW To discuss considerations of new paradigms for clinical drug development in pediatric oncology that incorporate our expanding knowledge and complexity of molecular alterations associated with cancer; advances in cancer immunology and cellular therapy; the increasing number of new anticancer drugs, therapeutic approaches, and potential combinations; and recent initiatives by regulatory agencies to improve access to safe and effective therapies. RECENT FINDINGS Cancer in children and adolescents is a rare event with significant long-term impact on individuals and society. Using multimodality therapy, stratified by patient and disease characteristics, the cure rate for childhood cancer exceeds 80%. Cancer genomics has transformed anticancer drug development. Understanding the genetic basis of pediatric cancers and the use of genomics for risk stratification has changed the focus of drug development from cytotoxic drugs to targeted therapeutic approaches. Advances in cancer immunology, immune checkpoint blockade, and cellular therapy offer novel approaches to harness T cells to treat cancer. To improve the outcome for children and adolescents with cancer and accelerate drug development, understanding drug and target interactions in preclinical models of pediatric cancer should be coupled with efficient clinical trial designs that incorporate biomarker selection, assessment of toxicity and drug exposure, and improved measures of response. SUMMARY Clinical trials for children and adolescents with cancer evaluate cytotoxic drugs, molecularly target drugs, immunotherapy as well as combination therapies. The framework for oncology clinical trials will continually adapt to improve efficiency of trials and evaluate new therapeutic approaches.
Collapse
|
27
|
Garralda E, Dienstmann R, Tabernero J. Pharmacokinetic/Pharmacodynamic Modeling for Drug Development in Oncology. Am Soc Clin Oncol Educ Book 2017; 37:210-215. [PMID: 28561730 DOI: 10.1200/edbk_180460] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
High drug attrition rates remain a critical issue in oncology drug development. A series of steps during drug development must be addressed to better understand the pharmacokinetic (PK) and pharmacodynamic (PD) properties of novel agents and, thus, increase their probability of success. As available data continues to expand in both volume and complexity, comprehensive integration of PK and PD information into a robust mathematical model represents a very useful tool throughout all stages of drug development. During the discovery phase, PK/PD models can be used to identify and select the best drug candidates, which helps characterize the mechanism of action and disease behavior of a given drug, to predict clinical response in humans, and to facilitate a better understanding about the potential clinical relevance of preclinical efficacy data. During early drug development, PK/PD modeling can optimize the design of clinical trials, guide the dose and regimen that should be tested further, help evaluate proof of mechanism in humans, anticipate the effect in certain subpopulations, and better predict drug-drug interactions; all of these effects could lead to a more efficient drug development process. Because of certain peculiarities of immunotherapies, such as PK and PD characteristics, PK/PD modeling could be particularly relevant and thus have an important impact on decision making during the development of these agents.
Collapse
Affiliation(s)
- Elena Garralda
- From the Early Drug Development Unit, Vall d'Hebron University Hospital and Vall d´Hebron Institute of Oncology, CIBERONC, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rodrigo Dienstmann
- From the Early Drug Development Unit, Vall d'Hebron University Hospital and Vall d´Hebron Institute of Oncology, CIBERONC, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josep Tabernero
- From the Early Drug Development Unit, Vall d'Hebron University Hospital and Vall d´Hebron Institute of Oncology, CIBERONC, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
28
|
Gore L, Ivy SP, Balis FM, Rubin E, Thornton K, Donoghue M, Roberts S, Bruinooge S, Ersek J, Goodman N, Schenkel C, Reaman G. Modernizing Clinical Trial Eligibility: Recommendations of the American Society of Clinical Oncology-Friends of Cancer Research Minimum Age Working Group. J Clin Oncol 2017; 35:3781-3787. [PMID: 28968169 DOI: 10.1200/jco.2017.74.4144] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Children have historically been excluded from first-in-human studies of promising new cancer drugs and later phase adult clinical trials. Delays in evaluation may result in off-label use without dosing information as the only access to new drugs. A multistakeholder workshop was convened in May 2016 by ASCO and Friends of Cancer Research to identify opportunities for when it would be scientifically appropriate to expand trial eligibility to include children younger than age 18 years in first-in-human and other adult cancer clinical trials. Methods This group convened experts from academia, government, and industry to review barriers to enrolling children and adolescents in oncology clinical trials. We evaluated the historical context, published literature, regulatory considerations, and myriad risks and benefits associated with lowering the age of enrollment on oncology clinical trials. Results We conclude that many of the historical concerns about including children early in oncology clinical trials do not apply in the current scientific and clinical environment of pediatric oncology and drug development; we provide specific recommendations for how the inclusion of children in early-phase investigational cancer drug trials might be accomplished. Automatic inclusion of pediatric patients is appropriate in early-phase trials that assess dose, safety, and pharmacokinetics in a variety of tumor types and later phase trials that assess efficacy in a specific disease that spans adult and pediatric populations. Conclusion Including children in appropriately designed adult clinical oncology trials is feasible and can be done in a way that enhances their access to these agents without compromising safety or development strategies.
Collapse
Affiliation(s)
- Lia Gore
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - S Percy Ivy
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Frank M Balis
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Eric Rubin
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Katherine Thornton
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Martha Donoghue
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Samantha Roberts
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Suanna Bruinooge
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Jennifer Ersek
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Nancy Goodman
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Caroline Schenkel
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| | - Gregory Reaman
- Lia Gore, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO; S. Percy Ivy, National Cancer Institute, Bethesda; Martha Donoghue and Gregory Reaman, US Food and Drug Administration, Silver Spring, MD; Frank M. Balis, Children's Hospital of Philadelphia, Philadelphia, PA; Eric Rubin, Merck Research Laboratories, Kenilworth, NJ; Katherine Thornton, Dana-Farber Cancer Institute, Boston, MA; Samantha Roberts, Friends of Cancer Research and Genentech; Nancy Goodman, Kids v Cancer, Washington, DC; Suanna Bruinooge and Caroline Schenkel, ASCO, Alexandria, VA; and Jennifer Ersek, Levine Cancer Institute, Charlotte, NC
| |
Collapse
|
29
|
Amoroso L, Haupt R, Garaventa A, Ponzoni M. Investigational drugs in phase II clinical trials for the treatment of neuroblastoma. Expert Opin Investig Drugs 2017; 26:1281-1293. [PMID: 28906153 DOI: 10.1080/13543784.2017.1380625] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Neuroblastoma (NB) is an embryonal tumor originating from undifferentiated neural crest cell, highly heterogeneous ranging from spontaneous regression to progression despite multimodal treatments. Approximately, 20% of patients are refractory to frontline therapy and 50% will relapse/progress after an initial response. The overall five year survival for high-risk neuroblastoma ranges from 35-45%. Despite enhanced understanding of NB biology and the addition of myeloablative chemotherapy, isotretinoin and immunotherapy, survival for high risk NB remains less than 50%. Areas covered: This review summarizes and gives a critical overview of phase II trials investigating therapies for relapsed-refractory and high risk neuroblastoma. Expert opinion: Several novel molecules have been developed and are currently under investigation for the treatment of NB. The trend of novel targeted agents is one towards individualized, tailored therapy, based on the molecular and biological differences that characterize tumors that seem similar based solely on histological analysis. The task of developing new molecules is particularly difficult for NB, given the recurrent development of new patterns of drug resistance. However, even if current research is focused towards identifying the best treatments for each children and young adult with a NB defined disease, a deeper knowledge of the molecular biology and genetics is needed.
Collapse
Affiliation(s)
- Loredana Amoroso
- a Department of Pediatric Oncology , Istituto G.Gaslini , Genova , Italy
| | - Riccardo Haupt
- b Epidemiology and Biostatistics Unit , Istituto G.Gaslini , Genova , Italy
| | - Alberto Garaventa
- a Department of Pediatric Oncology , Istituto G.Gaslini , Genova , Italy
| | - Mirco Ponzoni
- c Experimental Therapy Unit in Oncology , Istituto G. Gaslini , Genova , Italy
| |
Collapse
|