1
|
Pilbeam KL, Pradhan K, Croop J, Minard CG, Liu X, Voss SD, Isikwei E, Berg SL, Reid JM, Fox E, Weigel BJ. A phase 1 trial utilizing a pharmacokinetic endpoint to determine the optimal dose of ramucirumab in children and adolescents with relapsed or refractory solid tumors, including central nervous system tumors. Pediatr Blood Cancer 2024; 71:e30817. [PMID: 38189770 PMCID: PMC11220737 DOI: 10.1002/pbc.30817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/09/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Ramucirumab is a monoclonal antibody that binds the extracellular domain of vascular endothelial growth factor receptor (VEGFR-2) and prevents binding of VEGF ligands. Based on population pharmacokinetic (PK) analysis and correlation with efficacy in adults, a target steady state trough concentration (Css,min ) ≥ 50 µg/mL was established. PROCEDURES This phase 1 trial (ADVL1416) used a rolling six design and a PK primary endpoint to define the recommended phase 2 dose (RP2D) of ramucirumab in children with recurrent/refractory solid tumors. Two dose levels (DL) were planned (DL1: 8 mg/kg, DL2: 12 mg/kg administered intravenously [IV] every 2 weeks). Toxicity during the initial 6 weeks was used to assess maximum tolerated dose (MTD). Cycle 1 Day 42 trough (Cmin ) ≥ 50 µg/mL was the target concentration for the PK endpoint. At the RP2D, cohorts for PK expansion and children with central nervous tumors were planned. RESULTS Twenty-nine patients were enrolled; 28 were eligible; median age [range] = 13.5 [1-21] years; 22 were evaluable for the PK endpoint. Dose-limiting proteinuria occurred at both DLs; however, the MTD was not exceeded. At DL2 (12 mg/kg), the median Day 42 Cmin (n = 16) was 87.8 µg/mL; 15 of 16 patients achieved a Cmin ≥ 50 µg/mL. CONCLUSION Ramucirumab was well tolerated in children and adolescents with solid tumors. The RP2D for ramucirumab was 12 mg/kg IV every 2 weeks. This trial demonstrates the feasibility of incorporating a primary PK endpoint to determine dose escalation and the RP2D in children. Studies of ramucirumab in children with selected solid tumors are ongoing.
Collapse
Affiliation(s)
- Kristy L Pilbeam
- Spectrum Health, Pediatric Hematology Oncology, Helen DeVos Children's Hospital, Grand Rapids, Michigan, USA
| | | | - James Croop
- Pediatric Hematology Oncology, Riley Hospital for Children, Indianapolis, Indiana, USA
| | - Charles G Minard
- Baylor College of Medicine, Dan Duncan Cancer Institute, Houston, Texas, USA
| | - Xiaowei Liu
- Children's Oncology Group, Monrovia, California, USA
| | - Stephan D Voss
- Department Radiology, Dana-Farber/Harvard Cancer center, Boston, Massachusetts, USA
| | | | | | - Joel M Reid
- Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Elizabeth Fox
- Clinical Trials Administration, Saint Jude Children's Research Hospital Cancer Center, Memphis, Tennessee, USA
| | - Brenda J Weigel
- Pediatric Hematology Oncology, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| |
Collapse
|
2
|
A phase I study of irinotecan and temozolomide with bevacizumab in children with recurrent/refractory central nervous system tumors. Childs Nerv Syst 2022; 38:919-928. [PMID: 35260913 DOI: 10.1007/s00381-022-05479-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/26/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Children with relapsed/refractory central nervous system (CNS) tumors require novel combinations of therapies. Irinotecan and temozolomide (IT) is a frequently used therapy with an established toxicity profile. Bevacizumab is an anti-VEGF monoclonal antibody with demonstrated activity in CNS tumors. Therefore, the combination of these agents has therapeutic potential in CNS tumors. The objective of this study was to determine the maximum tolerated dose (MTD) of escalating dose IT combined with a fixed dose of bevacizumab (BIT) in children with relapsed/refractory CNS tumors. METHODS A phase I trial was performed in a 3 + 3 design. Therapy toxicities and radiologic responses to treatment were described. RESULTS One hundred eighty cycles of therapy were administered to 26 patients. The MTD of BIT was dose level 1, (bevacizumab 10 mg/kg on days 1 and 15, irinotecan 125 mg/m2 on days 1 and 15, and temozolomide 125 mg/m2 on days 1-5 of 28-day cycles). The regimen was well tolerated with primarily hematologic toxicity, which was not dose limiting. Among 22 response-evaluable patients, there was 1 complete response (CR), 6 partial responses (PR), and 10 stable diseases (SD) with an overall response rate (ORR: CR + PR) of 31.8%. CONCLUSION At the MTD, BIT therapy was well tolerated, and prolonged treatment courses of up to 24 cycles were feasible, with radiographic responses observed. Further evaluation is needed for efficacy in a phase II trial (NCT00876993, registered April 7, 2009, www. CLINICALTRIALS gov ).
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW The prognosis of pediatric patients with metastatic solid tumors remains poor, necessitating development of novel therapeutic strategies. The biology of the pediatric tumor microenvironment (TME) presents obstacles for the efficacy of current therapeutic approaches including immunotherapies. Targeting various aspects of the TME in pediatric patients with solid tumors represents a therapeutic opportunity that may improve outcomes. Here we will discuss recent advances in characterization of the TME, and clinical advances in targeting the immune, vascular, and stromal aspects of the TME. RECENT FINDINGS Although immunotherapies have shown limited success in the treatment of pediatric solid tumor patients thus far, optimization of these approaches to overcome the TME shows promise. In addition, there is increasing focus on the myeloid compartment as a therapeutic target. Vascular endothelial growth factor (VEGF) targeting has resulted in responses in some refractory pediatric solid tumors. There has been relatively little focus on stromal targeting; however, emerging preclinical data are improving our understanding of underlying biology, paving the way for future therapies. SUMMARY Although translation of TME-targeting therapies for pediatric solid tumors is in the early stages, we are optimistic that continued exploration of approaches aimed at rebalancing the TME will lead to improved outcomes for this population.
Collapse
Affiliation(s)
- Kristin M Wessel
- Tumor Microenvironment and Metastasis Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
4
|
Tsakatikas S, Papageorgiou G, Fioretzaki R, Kosmas C. An overview of current results with the vincristine-irinotecan-temozolomide combination with or without bevacizumab in pediatric, adolescence and adult solid tumors. Crit Rev Oncol Hematol 2021; 166:103457. [PMID: 34428555 DOI: 10.1016/j.critrevonc.2021.103457] [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: 10/17/2020] [Revised: 01/02/2021] [Accepted: 08/19/2021] [Indexed: 11/29/2022] Open
Abstract
Malignant tumors in young patients present a significant therapeutic challenge for physicians, partially due to their rarity and a relative lack of data, at least compared to adult tumors. As a result, there is an urgent need to explore new possible therapeutic regimens, either by introducing novel agents or by exploring combinations of existing agents. Vincristine, Temozolomide and Irinotecan are chemotherapeutic drugs which have emerged over the last six decades as monotherapy or as part of therapeutic regimens in various solid tumors. Combining these agents can yield strong synergistic effects, as suggested by preclinical data and results from clinical trials. Furthermore, adding novel molecules, such as anti-VEGF factor Bevacizumab to the aforementioned regimens, has shown efficacy in a limited number of trials, which are thoroughly analyzed throughout this review. Data presented throughout this paper suggest that VIT(b) regimen should be further explored in solid tumors in pediatric and adolescent patients.
Collapse
Affiliation(s)
- Sergios Tsakatikas
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| | - George Papageorgiou
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| | - Rodanthi Fioretzaki
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| | - Christos Kosmas
- Department of Medical Oncology & Hematopoietic Cell Transplant Unit, "Metaxa" Memorial Cancer Hospital, 18537, Piraeus, Greece.
| |
Collapse
|
5
|
Applebaum MA, Desai AV, Glade Bender JL, Cohn SL. Emerging and investigational therapies for neuroblastoma. Expert Opin Orphan Drugs 2017; 5:355-368. [PMID: 29062613 PMCID: PMC5649635 DOI: 10.1080/21678707.2017.1304212] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/06/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Treatment for children with clinically aggressive, high-risk neuroblastoma remains challenging. Less than 50% of patients with high-risk neuroblastoma will survive long-term with current therapies, and survivors are at risk for serious treatment-related late toxicities. Here, we review new and evolving treatments that may ultimately improve outcome for children with high-risk neuroblastoma with decreased potential for late adverse events. AREAS COVERED New strategies for treating high-risk neuroblastoma are reviewed including: radiotherapy, targeted cytotoxics, biologics, immunotherapy, and molecularly targeted agents. Recently completed and ongoing neuroblastoma clinical trials testing these novel treatments are highlighted. In addition, we discuss ongoing clinical trials designed to evaluate precision medicine approaches that target actionable somatic mutations and oncogenic cellular pathways. EXPERT OPINION Advances in genomic medicine and molecular biology have led to the development of early phase studies testing biologically rational therapies targeting aberrantly activated cellular pathways. Because many of these drugs have a wider therapeutic index than standard chemotherapeutic agents, these treatments may be more effective and less toxic than current strategies. However, to effectively integrate these targeted strategies, robust predictive biomarkers must be developed that will identify patients who will benefit from these approaches and rapidly match treatments to patients at diagnosis.
Collapse
Affiliation(s)
- Mark A. Applebaum
- Department of Pediatrics, University of Chicago, Chicago, Illinois, 60637, United States of America
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - Ami V. Desai
- Department of Pediatrics, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - Julia L. Glade Bender
- Department of Pediatrics, Columbia University Medical Center, New York, New York, 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, 10032
| | - Susan L. Cohn
- Department of Pediatrics, University of Chicago, Chicago, Illinois, 60637, United States of America
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois, 60637, United States of America
| |
Collapse
|
6
|
Zarei Mahmudabadi A, Masoomi Karimi M, Bahabadi M, Bagheri Hoseinabadi Z, JafariSani M, Ahmadi R. Inhibition of AGS Cancer Cell Proliferation following siRNA-Mediated Downregulation of VEGFR2. CELL JOURNAL 2016; 18:381-8. [PMID: 27602320 PMCID: PMC5011326 DOI: 10.22074/cellj.2016.4566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/24/2015] [Indexed: 12/03/2022]
Abstract
Objective Vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFRs) play
important roles in angiogenesis of different developmental mechanisms such as wound
healing, embryogenesis and diseases, including different types of cancer. VEGFR2 is
associated with cell proliferation, migration, and vascular permeability of endothelial cells.
Blocking VEGF and its receptors is suggested as a therapeutic approach to prevent tumor
growth. In this study, we aim to block VEGF signaling via small interfering RNA (siRNA)
inhibition of VEGFR2.
Materials and Methods In this experimental study, we used the RNA interference (RNAi)
mechanism to suppress expression of the VEGFR2 gene. We conducted the 3-(4,5-di-
methylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay, real-time polymerase
chain reaction (PCR), Western blot, and flow cytometry analyses of VEGFR2 expression.
Results Real-time PCR and Western blot results showed that VEGFR2 expression
significantly downregulated. This suppression was followed by inhibition of cell prolifera-
tion, reduction of viability, and induction of apoptosis in the cancer cells.
Conclusion These findings suggest that VEGFR2 has a role in cell proliferation and
tumor growth. Accordingly, it is suggested that VEGFR2 can be a therapeutic target
for controlling tumor growth and proliferation.
Collapse
Affiliation(s)
- Ali Zarei Mahmudabadi
- Department of Biochemical, Chemical Injuries Research Center, Baqiyatallah University of Medical Science, Tehran, Iran
| | - Masoomeh Masoomi Karimi
- Department of Immunology, Faculty of Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Majid Bahabadi
- Department of Biochemistry, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Moslem JafariSani
- Department of Basic Sciences, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Ahmadi
- Biochemistry Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
7
|
Jafari Sani M, Yazdi F, Masoomi Karimi M, Alizadeh J, Rahmati M, Zarei Mahmudabadi A. The siRNA-Mediated Down-Regulation of Vascular Endothelial Growth Factor Receptor1. IRANIAN RED CRESCENT MEDICAL JOURNAL 2016; 18:e23418. [PMID: 27275397 PMCID: PMC4893410 DOI: 10.5812/ircmj.23418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 04/16/2015] [Accepted: 06/08/2015] [Indexed: 11/24/2022]
Abstract
Background Angiogenesis is an important biological process involved in the proliferation of endothelial cells, tumor growth and metastasis. Vascular endothelial growth factor (VEGF) is considered as a prominent regulator of angiogenesis which exerts the aforementioned effect(s) through its respective receptors (VEGFR1 and VEGFR2). VEGF receptors are targeted as a therapeutic candidate for cancer growth inhibition. RNAi as a new and promising strategy has provided a useful means to specifically suppress gene expression in cancer cells. Objectives The current study aimed to down-regulate expression of the VEGFR1 using siRNA. Materials and Methods This experimental study designed specific siRNAs against VEGFR1. Total RNA was extracted from human umbilical vain endothelial cell (HUVEC) and subsequently cDNA was synthetized. PCR was performed using specific primers to amplify the target gene. After double digestion and purification, the gene was cloned into pEFGP-N1 expression vector. Then, AGS cells were transfected with recombinant pEGFP-N1 using lipofectamin. The gene expression and down-regulation were evaluated by fluorescence scanning, reverse transcription PCR (RT-PCR) and Western blot techniques. Results Fluorescent scanning, RT-PCR (27.68%) and western blot analysis (31.06%) showed that the expression of VEGFR1 was suppressed effectively. Conclusions The results of the current study showed that specifically designed siRNA can be considered as an appropriate strategy to suppress gene expression and might be a promising tool to prevent angiogenesis.
Collapse
Affiliation(s)
- Moslem Jafari Sani
- Biochemistry Department, School of Medicine, Shahroud University of Medical Sciences, Shahroud, IR Iran
| | - Foad Yazdi
- Biotechnology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Masoomeh Masoomi Karimi
- Immunology Department, School of Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, IR Iran
| | - Javad Alizadeh
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, IR Iran
| | - Majid Rahmati
- Biotechnology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Ali Zarei Mahmudabadi
- Biochemical Department, Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
- Corresponding Author: Ali Zarei Mahmudabadi, Biochemical Department, Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran. Tel: +98-9123277532, Fax: +98-2122830262, E-mail:
| |
Collapse
|
8
|
Millan NC, Poveda MJ, Cruz O, Mora J. Safety of bevacizumab in patients younger than 4 years of age. Clin Transl Oncol 2015; 18:464-8. [PMID: 26318053 DOI: 10.1007/s12094-015-1389-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/17/2015] [Indexed: 12/28/2022]
Abstract
PURPOSE Limited data exist regarding the safety and efficacy of bevacizumab in pediatric patients under the age of 4 years. Here, we report a large cohort of pediatric patients under 4 years of age treated with bevacizumab. METHODS The primary objective was to document adverse events with a possible relationship to bevacizumab. Patients (n = 16) were identified through retrospective chart review and harbored a variety of conditions (44% central nervous system (CNS) tumors, 31% vascular anomalies, 13% neuroblastoma, 12% other). RESULTS The median age was 34.3 months (range 4.9-47.3), including five patients <2 years of age. Patients received bevacizumab for a median duration of 6.2 months, alone or with chemotherapy, and a median dose of 9.25 mg/kg (range 7.0-11.8). Partial responses were seen in 19% of patients, and clinical improvements were seen in 69%. Adverse events known to be associated with bevacizumab occurred in 37%. Outcomes observed in this population resemble those reported for bevacizumab in older pediatric patients. The overall pattern and frequency of adverse events observed was similar to those seen in reports of older pediatric patients with a variety of conditions. The highest level of efficacy observed was seen among patients with vascular malformations or with low-grade CNS tumors. CONCLUSIONS Our results suggest that the use of bevacizumab is safe for the youngest children.
Collapse
Affiliation(s)
- N C Millan
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain
| | - M J Poveda
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain
| | - O Cruz
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain
| | - J Mora
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu de Barcelona, Passeig de Sant Joan de Déu num 2, Esplugues del Llobregat, 08950, Barcelona, Spain.
| |
Collapse
|
9
|
Abstract
Neuroblastoma (NB) is the third most common pediatric cancer. Although NB accounts for 7% of pediatric malignancies, it is responsible for more than 10% of childhood cancer-related mortality. Prognosis and treatment are determined by clinical and biological risk factors. Estimated 5-year survival rates for patients with non-high-risk and high-risk NB are more than 90% and less than 50%, respectively. Recent clinical trials have continued to reduce therapy for patients with non-high-risk NB, including the most favorable subsets who are often followed with observation approaches. In contrast, high-risk patients are treated aggressively with chemotherapy, radiation, surgery, and myeloablative and immunotherapies.
Collapse
|
10
|
Voss SD, Glade-Bender J, Spunt SL, DuBois SG, Widemann BC, Park JR, Leary SES, Nelson MD, Adamson PC, Blaney SM, Weigel B. Growth plate abnormalities in pediatric cancer patients undergoing phase 1 anti-angiogenic therapy: a report from the Children's Oncology Group Phase I Consortium. Pediatr Blood Cancer 2015; 62:45-51. [PMID: 25257751 PMCID: PMC4237627 DOI: 10.1002/pbc.25229] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/24/2014] [Indexed: 01/03/2023]
Abstract
BACKGROUND Pre-clinical studies suggest that anti-angiogenic agents may be toxic to the developing growth plate. The purpose of this study was to evaluate the incidence of growth plate abnormalities in children with refractory cancer undergoing anti-angiogenic therapy. PROCEDURE Targeted radiographic studies from 53 subjects enrolled on six separate Children's Oncology Group Phase 1 and Pilot Consortium clinical trials evaluating new anti-cancer agents interfering with angiogenesis were reviewed. Subjects received tyrosine kinase inhibitors with anti-angiogenic effects (n = 35), monoclonal antibodies targeting vascular endothelial growth factor (VEGF) (n = 13), or angiopoietin (n = 5). Radiographs of their distal femur/proximal tibia were obtained at baseline. Follow-up radiographs were obtained after odd-numbered treatment cycles in patients with open growth plates who did not experience disease progression prior to cycle 3. RESULTS Baseline and follow-up growth plate radiographs were acquired in 48/53 (90%) of patients. Five patients (9.4%), all of whom received a specific VEGF/VEGFR blocking agent (sunitinib [n = 1] or pazopanib [n = 4]), had growth plate abnormalities. Four patients had growth plate widening that was apparent on at least two successive radiographs, but was not confirmed by MRI. The fifth patient had progressive growth plate widening and evidence of physeal cartilage hypertrophy on MRI. Subsequent off treatment radiographs showed that the growth plate changes were reversible. CONCLUSION Growth plate abnormalities occur in a small, but relevant number of patients undergoing anti-angiogenic therapy. These results support the need for growth plate monitoring in children with open growth plates who are receiving anti-angiogenic therapy, and for improved methods to assess toxicity of anti-angiogenic agents to the developing skeleton.
Collapse
Affiliation(s)
- Stephan D. Voss
- Dept. of Radiology, Boston Children's Hospital; Dana Farber Cancer Institute Boston MA
| | | | - Sheri L. Spunt
- Lucile Packard Children's Hospital Stanford University, Pediatric Hematology/Oncology, Palo Alto CA
| | - Steven G. DuBois
- UCSF Medical Center-Parnassus, Pediatric Hematology/Oncology, San Francisco CA
| | - Brigitte C. Widemann
- Mark O Hatfield-Warren Grant Magnuson Clinical Center, Pharmacology & Experimental Therapeutics, Pediatric Oncology Branch, NCI, CCR, Bethesda MD
| | - Julie R. Park
- Seattle Children's Hospital, Hematology/Oncology, Seattle WA
| | | | | | - Peter C. Adamson
- Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia PA
| | - Susan M. Blaney
- Texas Children’s Cancer Center/Baylor College of Medicine, Houston, TX
| | - Brenda Weigel
- Division of Hematology and Oncology, University of Minnesota, Amplatz Children’s Hospital, Minneapolis, MN
| |
Collapse
|
11
|
Nella AA, Lodish MB, Fox E, Balis FM, Quezado MM, Whitcomb PO, Derdak J, Kebebew E, Widemann BC, Stratakis CA. Vandetanib successfully controls medullary thyroid cancer-related Cushing syndrome in an adolescent patient. J Clin Endocrinol Metab 2014; 99:3055-9. [PMID: 24617713 PMCID: PMC4154103 DOI: 10.1210/jc.2013-4340] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Ectopic Cushing syndrome due to ACTH secretion from metastatic medullary thyroid cancer (MTC) is associated with significant morbidity and mortality. OBJECTIVE The aim of the study was to describe the first case of Cushing syndrome associated with MTC in a pediatric patient and the successful reversal of Cushing syndrome with tyrosine kinase inhibitor (vandetanib) therapy. PATIENT AND METHODS A 17-year-old Brazilian adolescent presented with metastatic MTC and associated ACTH-dependent ectopic Cushing syndrome in the context of multiple endocrine neoplasia type 2B. When the patient was treated with the tyrosine kinase inhibitor vandetanib, rapid decrease in serum cortisol and improvement of clinical symptoms were observed. CONCLUSION We describe the first pediatric case of clinical and biochemical improvement of paraneoplastic MTC-related Cushing syndrome after treatment with vandetanib. Vandetanib and possibly other tyrosine kinase inhibitors may be a novel beneficial option in patients with neuroendocrine tumor-related ectopic Cushing syndrome.
Collapse
Affiliation(s)
- A A Nella
- Section on Endocrinology and Genetics (A.A.N., M.B.L., C.A.S.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892; Pharmacology and Experimental Therapeutics Section, Pediatric Oncology Branch (E.F., F.M.B., P.O.W., J.D., B.C.W.), General Surgical Pathology Section (M.M.Q.), and Endocrine Oncology Branch (E.K.), Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892; and Division of Oncology and Center for Childhood Cancer Research (E.F., F.M.B.), The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Smith MA. Lessons learned from adult clinical experience to inform evaluations of VEGF pathway inhibitors in children with cancer. Pediatr Blood Cancer 2014; 61:1497-505. [PMID: 24760743 DOI: 10.1002/pbc.25036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 02/27/2014] [Indexed: 01/07/2023]
Abstract
Agents targeting the vascular endothelial growth factor (VEGF) pathway have been studied in adults with cancer for nearly two decades. It is important to assess the lessons learned from this adult experience and to see how these lessons can help inform pediatric development of agents in this class. The benefit achieved from the use of VEGF pathway targeted agents for adult cancers has primarily been to delay for several months disease progression and less commonly time to death for conditions in which cure is not a reasonable expectation. VEGF pathway targeted agents have shown no efficacy when applied in the adjuvant setting. For adults with advanced cancer, prolongation of survival by 2-3 months is considered an important achievement in some settings. However, the primary goal of pediatric oncology clinical research is to identify treatments that allow children to be cured of their cancer and to grow to adulthood without treatment-induced limitations that lower their quality of survival. An important question for the pediatric oncology research community, pharmaceutical companies, and regulatory agencies to address in planning for future clinical trials is whether existing data support a role for VEGF pathway targeted agents in contributing to a therapeutic pathway to cure for children with cancer.
Collapse
|
13
|
Wagner L, Turpin B, Nagarajan R, Weiss B, Cripe T, Geller J. Pilot study of vincristine, oral irinotecan, and temozolomide (VOIT regimen) combined with bevacizumab in pediatric patients with recurrent solid tumors or brain tumors. Pediatr Blood Cancer 2013; 60:1447-51. [PMID: 23630159 DOI: 10.1002/pbc.24547] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/04/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND The combination of vincristine, oral irinotecan, and temozolomide (VOIT regimen) has shown antitumor activity in a pediatric Phase I trial. To further potentiate synergy, we assessed the safety and feasibility of adding bevacizumab to VOIT for children and young adults with recurrent tumors. METHODS Patients received vincristine (1.5 mg/m(2) on day 1), oral irinotecan (90 mg/m(2) on days 1-5), temozolomide (100-150 mg/m(2) on days 1-5), and bevacizumab (15 mg/kg on day 1) in 3-week cycles, which were repeated for up to six cycles. Cefixime prophylaxis was used to reduce irinotecan-associated diarrhea. RESULTS Thirteen patients received 36 total cycles. Six of the first 10 patients required dose reductions due to toxicity during the first cycle (n = 3) or subsequent cycles (n = 3), and these grade 3 side effects included prolonged nausea, dehydration, anorexia, neuropathy, diarrhea, and abdominal pain, as well as prolonged grade 4 neutropenia. After reducing daily temozolomide to 100 mg/m(2) , three additional patients tolerated therapy well without the need for dose reductions. Toxicities attributed to bevacizumab were limited to grade 1 epistaxis (1) and grade 2 proteinuria (1). Tumor responses were seen in both patients with Ewing sarcoma. CONCLUSIONS Reducing temozolomide from 150 to 100 mg/m(2) /day improved tolerability, and treatment with this lower temozolomide dose was feasible and convenient as outpatient therapy. Although responses were seen in Ewing sarcoma, the benefit of adding bevacizumab remains unclear.
Collapse
Affiliation(s)
- Lars Wagner
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Lexington, Kentucky, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Weiser DA, Kaste SC, Siegel MJ, Adamson PC. Imaging in childhood cancer: a Society for Pediatric Radiology and Children's Oncology Group Joint Task Force report. Pediatr Blood Cancer 2013; 60:1253-60. [PMID: 23572212 PMCID: PMC4636336 DOI: 10.1002/pbc.24533] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 02/21/2013] [Indexed: 12/25/2022]
Abstract
Contemporary medical imaging is a cornerstone of care for children with cancer. As 5-year survival rates for children with cancer exceed 80%, imaging technologies have evolved in parallel to include a wide array of modalities. Here, we overview the risks and benefits associated with commonly used imaging modalities and survey the current landscape of medical imaging for children with cancer. We find evidence-based imaging guidelines to assist in protocol development and to guide decision-making for optimal patient care are often lacking. The substantial variation in protocol-based recommendations for imaging both during and following therapy may hinder optimal clinical research and clinical care for children with cancer.
Collapse
Affiliation(s)
- Daniel A Weiser
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
15
|
Saha S, Islam MK, Shilpi JA, Hasan S. Inhibition of VEGF: a novel mechanism to control angiogenesis by Withania somnifera's key metabolite Withaferin A. In Silico Pharmacol 2013; 1:11. [PMID: 25505656 PMCID: PMC4230651 DOI: 10.1186/2193-9616-1-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/24/2013] [Indexed: 12/11/2022] Open
Abstract
Purpose Angiogenesis, or new blood vessel formation from existing one, plays both beneficial and detrimental roles in living organisms in different aspects. Vascular endothelial growth factor (VEGF), a signal protein, well established as key regulator of vasculogenesis and angiogenesis. VEGF ensures oxygen supply to the tissues when blood supply is not adequate, or tissue environment is in hypoxic condition. Limited expression of VEGF is necessary, but if it is over expressed, then it can lead to serious disease like cancer. Cancers that have ability to express VEGF are more efficient to grow and metastasize because solid cancers cannot grow larger than a limited size without adequate blood and oxygen supply. Anti-VEGF drugs are already available in the market to control angiogenesis, but they are often associated with severe side-effects like fetal bleeding and proteinuria in the large number of patients. To avoid such side-effects, new insight is required to find potential compounds as anti-VEGF from natural sources. In the present investigation, molecular docking studies were carried out to find the potentiality of Withaferin A, a key metabolite of Withania somnifera, as an inhibitor of VEGF. Methods Molecular Docking studies were performed in DockingServer and SwissDock. Bevacizumab, a commercial anti-VEGF drug, was used as reference to compare the activity of Withaferin A. X-ray crystallographic structure of VEGF, was retrieved from Protein Data Bank (PDB), and used as drug target protein. Structure of Withaferin A and Bevacizumab was obtained from PubChem and ZINC databases. Molecular visualization was performed using UCSF Chimera. Results Withaferin A showed favorable binding with VEGF with low binding energy in comparison to Bevacizumab. Molecular Docking studies also revealed potential protein-ligand interactions for both Withaferin A and Bevacizumab. Conclusions Conclusively our results strongly suggest that Withaferin A is a potent anti-VEGF agent as ascertained by its potential interaction with VEGF. This scientific hypothesis might provide a better insight to control angiogenesis as well as to control solid cancer growth and metastasis. Electronic supplementary material The online version of this article (doi:10.1186/2193-9616-1-11) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanjib Saha
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208 Bangladesh
| | - Md Khirul Islam
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208 Bangladesh
| | - Jamil A Shilpi
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208 Bangladesh ; Centre for Natural Products and Drug (CENAR), University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shihab Hasan
- School of Medicine, The University of Queensland (UQ), Brisbane, Queensland Australia ; Bioinformatics Lab, Queensland Institute of Medical Research (QIMR), Brisbane, Queensland Australia
| |
Collapse
|
16
|
Glade Bender JL, Lee A, Reid JM, Baruchel S, Roberts T, Voss SD, Wu B, Ahern CH, Ingle AM, Harris P, Weigel BJ, Blaney SM. Phase I pharmacokinetic and pharmacodynamic study of pazopanib in children with soft tissue sarcoma and other refractory solid tumors: a children's oncology group phase I consortium report. J Clin Oncol 2013; 31:3034-43. [PMID: 23857966 DOI: 10.1200/jco.2012.47.0914] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Pazopanib, an oral multikinase angiogenesis inhibitor, prolongs progression-free survival in adults with soft tissue sarcoma (STS). A phase I pharmacokinetic and pharmacodynamic study of two formulations of pazopanib was performed in children with STS or other refractory solid tumors. PATIENTS AND METHODS Pazopanib (tablet formulation) was administered once daily in 28-day cycles at four dose levels (275 to 600 mg/m(2)) using the rolling-six design. Dose determination for a powder suspension was initiated at 50% of the maximum-tolerated dose (MTD) for the intact tablet. Ten patients with STS underwent dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) scanning at baseline and 15 ± 2 days after initiation of pazopanib at the tablet MTD. RESULTS Fifty-three patients were enrolled; 51 were eligible (26 males; median age, 12.9 years; range, 3.8 to 23.9 years). Hematologic and nonhematologic toxicities were generally mild, with dose-limiting lipase, amylase, and ALT elevation, proteinuria, and hypertension. One patient with occult brain metastasis had grade 4 intracranial hemorrhage. The MTD was 450 mg/m(2) for tablet and 160 mg/m(2) for suspension. Steady-state trough concentrations were reached by day 15 and did not seem to be dose dependent. One patient each with hepatoblastoma or desmoplastic small round cell tumor achieved a partial response; eight patients had stable disease for ≥ six cycles, seven of whom had sarcoma. All patients with evaluable DCE-MRI (n = 8) experienced decreases in tumor blood volume and permeability (P < .01). Placental growth factor increased, whereas endoglin and soluble vascular endothelial growth factor receptor-2 decreased (P < .01; n = 41). CONCLUSION Pazopanib is well tolerated in children, with evidence of antiangiogenic effect and potential clinical benefit in pediatric sarcoma.
Collapse
|
17
|
Abstract
Abstract
Angiogenesis plays a pivotal role in the growth and metastasis of adult and pediatric solid tumors. Clinical investigation of angiogenesis inhibitors is currently under way for childhood cancers. While the pediatric study of aflibercept provides a proof-of-principle, challenges remain in developing clinical endpoints and biomarkers of angiogenesis for pediatric trials. Clin Cancer Res; 18(18); 4868–71. ©2012 AACR.
Collapse
Affiliation(s)
- Cindy H. Chau
- Authors' Affiliation: Molecular Pharmacology Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - William D. Figg
- Authors' Affiliation: Molecular Pharmacology Section, Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|