1
|
Bergamaschi L, Chiaravalli S, Signoroni S, Di Bartolomeo M, Ferrari A. Management and pharmacotherapy of pediatric colorectal carcinoma: a review. Expert Opin Pharmacother 2023; 24:1527-1535. [PMID: 37358925 DOI: 10.1080/14656566.2023.2230123] [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: 04/05/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION Colorectal carcinoma (CRC) is one of the most common tumors in adult, but is extremely rare in children. In childhood, CRC often presents unfavorable aggressive histotypes, advanced clinical stage at onset and a worse prognosis. Pediatric CRC series are limited and include few patients, therefore information about treatment strategy and pharmacotherapy is scarce. For this reason, management of these patients represents a real challenge for pediatric oncologists. AREAS COVERED The authors provide an overview of the general features and management strategies of pediatric CRC with specific attention to systemic treatment. Literature data regarding pharmacotherapy in published pediatric series are summarized and analyzed in detail, according to adult treatment standards. EXPERT OPINION In the absence of specific recommendations for pediatric CRC, the general therapeutic strategy should follow the same principles as for adults and should be the result of a multidisciplinary discussion. Patient access to optimal treatment is difficult due to the lack of new drugs approved for the pediatric age group and non-availability of clinical trials. Collaboration between pediatric and adult oncologists is considered crucial in order to overcome these issues and find solutions to increase knowledge and improve the outcome of such a rare disease in children.
Collapse
Affiliation(s)
- Luca Bergamaschi
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Stefano Chiaravalli
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Stefano Signoroni
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Maria Di Bartolomeo
- Gastrointestinal Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
2
|
Tang Y, Gutmann DH. Neurofibromatosis Type 1-Associated Optic Pathway Gliomas: Current Challenges and Future Prospects. Cancer Manag Res 2023; 15:667-681. [PMID: 37465080 PMCID: PMC10351533 DOI: 10.2147/cmar.s362678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
Optic pathway glioma (OPG) occurs in as many as one-fifth of individuals with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Generally considered low-grade and slow growing, many children with NF1-OPGs remain asymptomatic. However, due to their location within the optic pathway, ~20-30% of those harboring NF1-OPGs will experience symptoms, including progressive vision loss, proptosis, diplopia, and precocious puberty. While treatment with conventional chemotherapy is largely effective at attenuating tumor growth, it is not clear whether there is much long-term recovery of visual function. Additionally, because these tumors predominantly affect young children, there are unique challenges to NF1-OPG diagnosis, monitoring, and longitudinal management. Over the past two decades, the employment of authenticated genetically engineered Nf1-OPG mouse models have provided key insights into the function of the NF1 protein, neurofibromin, as well as the molecular and cellular pathways that contribute to optic gliomagenesis. Findings from these studies have resulted in the identification of new molecular targets whose inhibition blocks murine Nf1-OPG growth in preclinical studies. Some of these promising compounds have now entered into early clinical trials. Future research focused on defining the determinants that underlie optic glioma initiation, expansion, and tumor-induced optic nerve injury will pave the way to personalized risk assessment strategies, improved tumor monitoring, and optimized treatment plans for children with NF1-OPG.
Collapse
Affiliation(s)
- Yunshuo Tang
- Department of Ophthalmology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Watanabe T, Mizuno HL, Norimatsu J, Obara T, Cabral H, Tsumoto K, Nakakido M, Kawauchi D, Anraku Y. Ligand Installation to Polymeric Micelles for Pediatric Brain Tumor Targeting. Polymers (Basel) 2023; 15:polym15071808. [PMID: 37050422 PMCID: PMC10097392 DOI: 10.3390/polym15071808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Medulloblastoma is a life-threatening disease with poor therapeutic outcomes. In chemotherapy, low drug accumulation has been a cause of these outcomes. Such inadequate response to treatments has been associated with low drug accumulation, particularly with a limited cellular uptake of drugs. Recently, the conjugation of drugs to ligand molecules with high affinity to tumor cells has attracted much attention for enhancing drug internalization into target cells. Moreover, combining tumor-targeting ligands with nano-scaled drug carriers can potentially improve drug loading capacity and the versatility of the delivery. Herein, we focused on the possibility of targeting CD276/B7-H3, which is highly expressed on the medulloblastoma cell membrane, as a strategy for enhancing the cellular uptake of ligand-installed nanocarriers. Thus, anti-CD276 antibodies were conjugated on the surface of model nanocarriers based on polyion complex micelles (PIC/m) via click chemistry. The results showed that the anti-CD276 antibody-installed PIC/m improved intracellular delivery into CD276-expressing medulloblastoma cells in a CD276-dependent manner. Moreover, increasing the number of antibodies on the surface of micelles improved the cellular uptake efficiency. These observations indicate the potential of anti-CD276 antibody-installed nanocarriers for promoting drug delivery in medulloblastoma.
Collapse
Affiliation(s)
- Takayoshi Watanabe
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hayato Laurence Mizuno
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-8551, Japan
| | - Jumpei Norimatsu
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Takumi Obara
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo 113-8654, Japan
| | - Makoto Nakakido
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Daisuke Kawauchi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-8551, Japan
| | - Yasutaka Anraku
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan
| |
Collapse
|
4
|
Camero S, Cassandri M, Pomella S, Milazzo L, Vulcano F, Porrazzo A, Barillari G, Marchese C, Codenotti S, Tomaciello M, Rota R, Fanzani A, Megiorni F, Marampon F. Radioresistance in rhabdomyosarcomas: Much more than a question of dose. Front Oncol 2022; 12:1016894. [PMID: 36248991 PMCID: PMC9559533 DOI: 10.3389/fonc.2022.1016894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/12/2022] [Indexed: 11/15/2022] Open
Abstract
Management of rhabdomyosarcoma (RMS), the most common soft tissue sarcoma in children, frequently accounting the genitourinary tract is complex and requires a multimodal therapy. In particular, as a consequence of the advancement in dose conformity technology, radiation therapy (RT) has now become the standard therapeutic option for patients with RMS. In the clinical practice, dose and timing of RT are adjusted on the basis of patients' risk stratification to reduce late toxicity and side effects on normal tissues. However, despite the substantial improvement in cure rates, local failure and recurrence frequently occur. In this review, we summarize the general principles of the treatment of RMS, focusing on RT, and the main molecular pathways and specific proteins involved into radioresistance in RMS tumors. Specifically, we focused on DNA damage/repair, reactive oxygen species, cancer stem cells, and epigenetic modifications that have been reported in the context of RMS neoplasia in both in vitro and in vivo studies. The precise elucidation of the radioresistance-related molecular mechanisms is of pivotal importance to set up new more effective and tolerable combined therapeutic approaches that can radiosensitize cancer cells to finally ameliorate the overall survival of patients with RMS, especially for the most aggressive subtypes.
Collapse
Affiliation(s)
- Simona Camero
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Cassandri
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
- Department of Oncohematology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Silvia Pomella
- Department of Oncohematology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luisa Milazzo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Vulcano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Porrazzo
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
- Units of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Silvia Codenotti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Miriam Tomaciello
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Rossella Rota
- Department of Oncohematology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Francesco Marampon
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Saker Z, Rizk M, Bahmad HF, Nabha SM. Targeting Angiogenic Factors for the Treatment of Medulloblastoma. Curr Treat Options Oncol 2022; 23:864-886. [PMID: 35412196 DOI: 10.1007/s11864-022-00981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2022] [Indexed: 11/24/2022]
Abstract
OPINION STATEMENT Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.
Collapse
Affiliation(s)
- Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mahdi Rizk
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA.
| | - Sanaa M Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
6
|
Evans M, Gill R, Bull KS. Does a Bevacizumab-based regime have a role in the treatment of children with diffuse intrinsic pontine glioma? A systematic review. Neurooncol Adv 2022; 4:vdac100. [PMID: 35821674 PMCID: PMC9270727 DOI: 10.1093/noajnl/vdac100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background There are no effective treatments for diffuse intrinsic pontine glioma (DIPG); median survival is 11.2 months. Bevacizumab has the potential to improve quality of life (QOL) and survival in DIPG but has never been evaluated systematically. The aim of this review was to assess Bevacizumab’s role in the treatment of DIPG. Methods MEDLINE, EMBASE, Scopus, and Web of Science were searched for relevant studies using terms developed from alternatives for Bevacizumab and DIPG. One reviewer screened titles and abstracts, then two reviewers screened full texts. Data were extracted into tables and quality assessed using methodological index for non-randomized studies and JBI tools. Results Searching revealed 1001 papers; after deduplication 851 remained. After screening of titles and abstracts, then 28 full texts, 11 studies were included. Four studies reported a median overall survival longer than historical data, however, two found no significant impact of Bevacizumab. Five studies reported a radiological response in a proportion of participants and two reported no response. Three studies, evaluating clinical response, reported improvement in a proportion of patients. Three studies, evaluating QOL, reported stability or improvement. Four studies, evaluating steroid use, reported reductions in the proportion of patients receiving steroids. In radiation necrosis treatment, Bevacizumab led to clinical improvement in 6/12 patients in 2 studies and permitted a reduction in steroid use in most patients. Conclusions Insufficient evidence means the role of Bevacizumab in the treatment of DIPG is unclear. However, Bevacizumab may be beneficial to some patients. The review highlights the need for further research in this area.
Collapse
Affiliation(s)
- Mia Evans
- Faculty of medicine, University of Southampton , Southampton , UK
| | - Ria Gill
- Faculty of medicine, University of Southampton , Southampton , UK
| | - Kim S Bull
- Clinical and Experimental Sciences, University of Southampton , Southampton , UK
| |
Collapse
|
7
|
Giotta Lucifero A, Elbabaa SK, Baldoncini M, Bruno N, Savasta S, Marseglia GL, Luzzi S. Novel "T-Dimension" Therapies for Pediatric Optic Pathway Glioma: A Timely, Targeted, and Tailored Treatment Trend. Pediatr Neurosurg 2022; 57:161-174. [PMID: 35588700 DOI: 10.1159/000524873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 04/26/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Novel targeted and tailored therapies can substantially improve the prognosis for optic pathway glioma (OPG), especially when implemented in a timely manner. However, their tremendous potential remains underestimated. Therefore, in this study, we provide an updated overview of the clinical trials, current trends, and future perspectives for OPG's novel therapeutic strategies. METHODS We completed an extensive literature review using the PubMed, MEDLINE, and ClinicalTrials.gov databases. We analyzed and reported the data following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS Thioguanine, procarbazine, lomustine, and vincristine/vinblastine, as well as cisplatin-etoposide, provided excellent results in advanced-phase trials. Selumetinib and trametinib, two oral MEK inhibitors, have been approved for recurrent or refractory OPGs in association with the angiogenetic inhibitor bevacizumab. Among the mTOR inhibitors, everolimus and sirolimus showed the best results. Stereotactic radiosurgery and proton beam radiation therapy have advantages over conventional radiotherapy regimens. Timely treatment is imperative for acute visual symptoms with evidence of tumor progression. This latest evidence can help define a novel "T-Dimension" for pediatric OPG therapies. CONCLUSION The novel "T-Dimension" for pediatric OPGs is based on recent evidence-based treatments, including combination chemotherapy regimens, molecular targeted therapies, stereotactic radiosurgery, and proton beam radiation therapy. Additional clinical trials are essential for validating each of these new therapies.
Collapse
Affiliation(s)
- Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Samer K Elbabaa
- Department of Pediatric Neurosurgery, Leon Pediatric Neuroscience Center of Excellence, Arnold Palmer Hospital for Children, Orlando, Florida, USA
| | - Matias Baldoncini
- Laboratory of Neuroanatomic Microsurgical-LaNeMic-II Division of Anatomy, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Nunzio Bruno
- Division of Neurosurgery, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy
| | - Salvatore Savasta
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Gian Luigi Marseglia
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
8
|
Shaik S, Maegawa S, Gopalakrishnan V. Medulloblastoma: novel insights into emerging therapeutic targets. Expert Opin Ther Targets 2021; 25:615-619. [PMID: 34602009 DOI: 10.1080/14728222.2021.1982896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Shavali Shaik
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shinji Maegawa
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vidya Gopalakrishnan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Poli E, Cattelan M, Zanetti I, Scagnellato A, Giordano G, Zin A, Bisogno G, Bonvini P. Autoantibody profiling of alveolar rhabdomyosarcoma patients unveils tumor-associated antigens with diagnostic and prognostic significance. Oncoimmunology 2021; 10:1954765. [PMID: 34367733 PMCID: PMC8312597 DOI: 10.1080/2162402x.2021.1954765] [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] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alveolar rhabdomyosarcoma (ARMS) is a highly aggressive subtype of childhood cancer for which efficacious treatments are needed. Immunotherapy represents a new therapeutic opportunity to pursue, but it requires the identification of worthwhile tumor antigens. Herein, we exploited the capacity of ARMS autoantibodies to recognize tumor self-antigens, probing human protein microarrays with plasma from ARMS patients and healthy subjects. We assessed the autoantibody response in ARMS, validated data with independent techniques, and estimated autoantibodies diagnostic and prognostic significance by receiver-operator characteristic curves (ROC), uni- and multivariate analysis. Of the 48 tumor antigens identified, General Transcription Factor II-I (GTF2i) and Protocadherin Gamma Subfamily C5 (PCDHGC5) were selected as candidate targets to validate tumor-restricted antigen expression and autoantibody reactivity through an independent technique and wider cohort of cases. GTF2i and PCDHGC5 overexpression was observed in tumor tissues compared to normal counterparts, and anti-GTF2i and -PCDHGC5 autoantibodies were found able to distinguish ARMS patients from healthy subjects as well as cases with different histology. Moreover, low levels of PCDHGC5 autoantibodies characterized patients with worse event-free survival and proved to be an independent negative prognostic factor. This approach provided the first comprehensive autoantibody profile of ARMS, gave novel insights into the immune response of this malignancy and paved the way toward novel potential antibody-based therapeutic applications suitable to improve the survival of ARMS patients.
Collapse
Affiliation(s)
- Elena Poli
- Department of Woman's and Children's Health Hematology and Oncology Unit, University of Padua, Padua, Italy
| | - Manuela Cattelan
- Department of Statistical Sciences, University of Padua, Padua, Italy
| | - Ilaria Zanetti
- Department of Woman's and Children's Health Hematology and Oncology Unit, University of Padua, Padua, Italy
| | - Angela Scagnellato
- Department of Woman's and Children's Health Hematology and Oncology Unit, University of Padua, Padua, Italy
| | - Giuseppe Giordano
- Department of Woman's and Children's Health Hematology and Oncology Unit, University of Padua, Padua, Italy.,Institute of Pediatric Research (IRP), Fondazione Città Della Speranza, Padua, Italy
| | - Angelica Zin
- Institute of Pediatric Research (IRP), Fondazione Città Della Speranza, Padua, Italy
| | - Gianni Bisogno
- Department of Woman's and Children's Health Hematology and Oncology Unit, University of Padua, Padua, Italy
| | - Paolo Bonvini
- Institute of Pediatric Research (IRP), Fondazione Città Della Speranza, Padua, Italy
| |
Collapse
|
10
|
Ollauri-Ibáñez C, Astigarraga I. Use of Antiangiogenic Therapies in Pediatric Solid Tumors. Cancers (Basel) 2021; 13:E253. [PMID: 33445470 PMCID: PMC7827326 DOI: 10.3390/cancers13020253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cancer is an important cause of death in childhood. In recent years, scientists have made an important effort to achieve greater precision and more personalized treatments against cancer. But since only a few pediatric patients have identifiable therapeutic targets, other ways to stop the neoplastic cell proliferation and dissemination are needed. Therefore, the inhibition of general processes involved in the growth and behavior of tumors can be a relevant strategy for the development of new cancer therapies. In the case of solid tumors, one of these processes is angiogenesis, essential for tumor growth and generation of metastases. This review summarizes the results obtained with the use of antiangiogenic drugs in the main pediatric malignant solid tumors and also an overview of clinical trials currently underway. It should be noted that due to the rarity and heterogeneity of the different types of pediatric cancer, most studies on antiangiogenic drugs include only a small number of patients or isolated clinical cases, so they are not conclusive and further studies are needed.
Collapse
Affiliation(s)
- Claudia Ollauri-Ibáñez
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
| | - Itziar Astigarraga
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
- Pediatrics Department, Hospital Universitario Cruces, 48903 Barakaldo, Spain
- Pediatrics Department, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| |
Collapse
|
11
|
Koo J, Hayashi M, Verneris MR, Lee-Sherick AB. Targeting Tumor-Associated Macrophages in the Pediatric Sarcoma Tumor Microenvironment. Front Oncol 2020; 10:581107. [PMID: 33381449 PMCID: PMC7769312 DOI: 10.3389/fonc.2020.581107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
For many pediatric sarcoma patients, multi-modal therapy including chemotherapy, radiation, and surgery is sufficient to cure their disease. However, event-free and overall survival rates for patients with more advanced disease are grim, necessitating the development of novel therapeutic approaches. Within many pediatric sarcomas, the normal immune response, including recognition and destruction of cancer cells, is lost due to the highly immune suppressive tumor microenvironment (TME). In this setting, tumor cells evade immune detection and capitalize on the immune suppressed microenvironment, leading to unchecked proliferation and metastasis. Recent preclinical and clinical approaches are aimed at understanding this immune suppressive microenvironment and employing cancer immunotherapy in an attempt to overcome this, by renewing the ability of the immune system to recognize and destroy cancer cells. While there are several factors that drive the attenuation of immune responses in the sarcoma TME, one of the most remarkable are tumor associated macrophage (TAMs). TAMs suppress immune cytolytic function, promote tumor growth and metastases, and are generally associated with a poor prognosis in most pediatric sarcoma subtypes. In this review, we summarize the mechanisms underlying TAM-facilitated immune evasion and tumorigenesis and discuss the potential therapeutic application of TAM-focused drugs in the treatment of pediatric sarcomas.
Collapse
Affiliation(s)
- Jane Koo
- Department of Pediatric Hematology/Oncology/Bone Marrow Transplant, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, United States
| | - Masanori Hayashi
- Department of Pediatric Hematology/Oncology/Bone Marrow Transplant, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, United States
| | - Michael R Verneris
- Department of Pediatric Hematology/Oncology/Bone Marrow Transplant, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, United States
| | - Alisa B Lee-Sherick
- Department of Pediatric Hematology/Oncology/Bone Marrow Transplant, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
12
|
Russo I, Di Paolo V, Crocoli A, Mastronuzzi A, Serra A, Di Paolo PL, Di Giannatale A, Miele E, Milano GM. A Chart Review on the Feasibility and Safety of the Vincristine Irinotecan Pazopanib (VIPaz) Association in Children and Adolescents With Resistant or Relapsed Sarcomas. Front Oncol 2020; 10:1228. [PMID: 32850366 PMCID: PMC7425124 DOI: 10.3389/fonc.2020.01228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/16/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Pediatric patients with relapsed or refractory sarcomas have poor outcome and need novel therapies that provide disease control while maintaining an acceptable quality of life. The safety of vincristine, irinotecan, and pazopanib (VIPaz) association has not yet been published in this population. Methods: A chart review was conducted in children and adolescents with relapsed or refractory bone and soft tissue sarcomas who received VIPaz in our institution. Results: One hundred sixty-six patients with a diagnosis of soft or bone sarcoma were admitted to our hospital in the period between March 2015 and August 2018, 30 were relapsed or resistant. Seventeen out of 30 resistant or relapsed patients (median age, 14 years) received 114 VIPaz cycles (median six cycles per patient, range 1-17). Sixteen courses (15%) resulted in gastrointestinal toxicity with Grade two diarrhea; 35 courses (30%) resulted in Grade ≥3 neutropenia. One patient presented Grade two hypothyroidism after nine courses, and another one had Grade two hyperbilirubinemia after 12 courses. Two and five patients required a 25% dose reduction of irinotecan (because of diarrhea) and pazopanib (because of neutropenia four and hyperbilirubinemia 1), respectively. No patient experienced heart failure, hypertension, nor posterior reversible encephalopathy syndrome. Pneumothorax was not reported in any case even in lung metastatic patients. After two and four VIPaz cycles, we observed one complete response (CR), five partial responses (PRs), seven stable diseases (SDs), and four progressive diseases (PDs). With a median follow-up of 15 months (range 3-32), five out of 17 (29%) patients were alive, and four patients were in continuous CR after 12 VIPaz cycles. Conclusions: The VIPaz regimen might be a safe option in children and adolescents with relapsed or refractory sarcomas otherwise unable to be enrolled in other clinical trials; on the other hand, the efficacy of pazopanib observed cannot be sustained from the current study.
Collapse
Affiliation(s)
- Ida Russo
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Virginia Di Paolo
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessandro Crocoli
- Department of Surgery - Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Angela Mastronuzzi
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Annalisa Serra
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Pier Luigi Di Paolo
- Department of Radiology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Angela Di Giannatale
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Evelina Miele
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giuseppe Maria Milano
- Department of Pediatric Hematology/Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
13
|
Miwa S, Yamamoto N, Hayashi K, Takeuchi A, Igarashi K, Tsuchiya H. Recent Advances and Challenges in the Treatment of Rhabdomyosarcoma. Cancers (Basel) 2020; 12:cancers12071758. [PMID: 32630642 PMCID: PMC7409313 DOI: 10.3390/cancers12071758] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
Rhabdomyosarcoma, the most common soft tissue sarcoma noted in childhood, requires multimodality treatment, including chemotherapy, surgical resection, and/or radiation therapy. The majority of the patients with localized rhabdomyosarcoma can be cured; however, the long-term outcomes in patients with metastatic rhabdomyosarcoma remain poor. The standard chemotherapy regimen for patients with rhabdomyosarcoma is the combination of vincristine, actinomycin, and cyclophosphamide/ifosfamide. In recent clinical trials, modifications of the standard chemotherapy protocol have shown improvements in the outcomes in patients with rhabdomyosarcoma. In various type of malignancies, new treatments, such as molecular targeted drugs and immunotherapies, have shown superior clinical outcomes compared to those of standard treatments. Therefore, it is necessary to assess the benefits of these treatments in patients with rhabdomyosarcoma. Moreover, recent basic and clinical studies on rhabdomyosarcoma have reported promising therapeutic targets and novel therapeutic approaches. This article reviews the recent challenges and advances in the management of rhabdomyosarcoma.
Collapse
|
14
|
Xu Y, Li Q, Ma H, Sun T, Xiang R, Di F. Therapeutic effect and side effects of Bevacizumab combined with Irinotecan in the treatment of paediatric intracranial tumours: Meta-analysis and Systematic Review. J Clin Pharm Ther 2020; 45:1363-1371. [PMID: 32598559 PMCID: PMC7689821 DOI: 10.1111/jcpt.13228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/19/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Bevacizumab (BVZ) is an angiogenesis inhibitor that often works well with chemotherapeutic drugs for the treatment of solid intracranial tumours in children. This meta-analysis discusses the efficacy and side effects of BVZ combined with irinotecan in the treatment of patients (younger than 21 years of age) with recurrent, progressive or refractory intracranial tumours. METHODS We searched for articles published before 31 October 2018 in PubMed, EMBASE, Cochrane library and Web of Science. We selected relevant literature on the combination of BVZ and irinotecan in the treatment of children with intracranial tumours. Objective response was evaluated by combining partial response (PR), complete response (CR), stable disease (SD) and progressive disease (PD), and survival time was evaluated by combining overall survival (OS) and progression-free survival (PFS); common side effects were also analysed. All data included were obtained from single-arm data, with no control groups. RESULTS AND DISCUSSION A total of 13 studies involving 272 patients were included. We found that out of 41% patients who showed an objective response following the BVZ therapy combined with irinotecan, 28% achieved a PR, 13% achieved a CR, 32% showed a SD, and 43% had a PD; PFS and OS were 6.47 and 11.9 months, respectively; gastrointestinal dysfunction, leukopenia and hypertension were the three most common adverse events, accounting for 36.7%, 33.6% and 22.1%, respectively, whereas musculoskeletal disorders had the lowest occurrence, accounting for 3.9%. WHAT IS NEW AND CONCLUSION BVZ combined with irinotecan-based chemotherapy had a better response and prolonged survival in the treatment of paediatric intracranial tumours than radiation therapy or chemotherapy. Gastrointestinal dysfunction, leukopenia and hypertension were the toxic side effects with the highest incidence.
Collapse
Affiliation(s)
- Yan Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qiang Li
- Department of NeurosurgeryXinle City HospitalXinleHebeiChina
| | - Hai‐Yang Ma
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Tao Sun
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Ruo‐Lan Xiang
- Department of Physiology and PathophysiologyPeking University School of Basic Medical SciencesBeijingChina
| | - Fei Di
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryZhangJiakou First HospitalZhangjiakouHebeiChina
| |
Collapse
|
15
|
Yamasaki F, Takano M, Yonezawa U, Taguchi A, Kolakshyapati M, Okumichi H, Kiuchi Y, Kurisu K. Bevacizumab for optic pathway glioma with worsening visual field in absence of imaging progression: 2 case reports and literature review. Childs Nerv Syst 2020; 36:635-639. [PMID: 31701281 DOI: 10.1007/s00381-019-04407-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/03/2019] [Indexed: 11/27/2022]
Abstract
Children with optic pathway gliomas (OPGs) frequently suffer from problems of visual function resulting from tumors. Previous reports showed that bevacizumab improved visual function in patients with OPG via tumor response to treatment. In these two case reports, we show that bevacizumab improved visual field without tumor response as seen in imaging. Both, a 10-year-old girl and a 6-year-old boy, had previous history of treatment with platinum-based chemotherapy. They had visual deterioration without tumor progression on MR imaging. Bevacizumab effectively and immediately improved visual field in both patients without imaging response of OPG. We emphasize that bevacizumab should be considered for patients with OPGs having visual deterioration without tumor progression.
Collapse
Affiliation(s)
- Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Motoki Takano
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ushio Yonezawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Akira Taguchi
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Manish Kolakshyapati
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Department of Neuroscience, B&B Hospital, Gwarko, Lalitpur, Nepal
| | - Hideaki Okumichi
- Department of Ophthalmology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yoshiaki Kiuchi
- Department of Ophthalmology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
16
|
Abstract
As a cancer predisposition syndrome, individuals with neurofibromatosis type 1 (NF1) are at increased risk for the development of both benign and malignant tumors. One of the most common locations for these cancers is the central nervous system, where low-grade gliomas predominate in children. During early childhood, gliomas affecting the optic pathway are most frequently encountered, whereas gliomas of the brainstem and other locations are observed in slightly older children. In contrast, the majority of gliomas arising in adults with NF1 are malignant cancers, typically glioblastoma, involving the cerebral hemispheres. Our understanding of the pathogenesis of NF1-associated gliomas has been significantly advanced through the use of genetically engineered mice, yielding new targets for therapeutic drug design and evaluation. In addition, Nf1 murine glioma models have served as instructive platforms for defining the cell of origin of these tumors, elucidating the critical role of the tumor microenvironment in determining tumor growth and vision loss, and determining how cancer risk factors (sex, germline NF1 mutation) impact on glioma formation and progression. Moreover, these preclinical models have permitted early phase analysis of promising drugs that reduce tumor growth and attenuate vision loss, as an initial step prior to translation to human clinical trials.
Collapse
Affiliation(s)
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
17
|
Bautista F, Fioravantti V, de Rojas T, Carceller F, Madero L, Lassaletta A, Moreno L. Medulloblastoma in children and adolescents: a systematic review of contemporary phase I and II clinical trials and biology update. Cancer Med 2017; 6:2606-2624. [PMID: 28980418 PMCID: PMC5673921 DOI: 10.1002/cam4.1171] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022] Open
Abstract
Survival rates for patients with medulloblastoma have improved in the last decades but for those who relapse outcome is dismal and new approaches are needed. Emerging drugs have been tested in the last two decades within the context of phase I/II trials. In parallel, advances in genetic profiling have permitted to identify key molecular alterations for which new strategies are being developed. We performed a systematic review focused on the design and outcome of early-phase trials evaluating new agents in patients with relapsed medulloblastoma. PubMed, clinicaltrials.gov, and references from selected studies were screened to identify phase I/II studies with reported results between 2000 and 2015 including patients with medulloblastoma aged <18 years. A total of 718 studies were reviewed and 78 satisfied eligibility criteria. Of those, 69% were phase I; 31% phase II. Half evaluated conventional chemotherapeutics and 35% targeted agents. Overall, 662 patients with medulloblastoma/primitive neuroectodermal tumors were included. The study designs and the response assessments were heterogeneous, limiting the comparisons among trials and the correct identification of active drugs. Median (range) objective response rate (ORR) for patients with medulloblastoma in phase I/II studies was 0% (0-100) and 6.5% (0-50), respectively. Temozolomide containing regimens had a median ORR of 16.5% (0-100). Smoothened inhibitors trials had a median ORR of 8% (3-8). Novel drugs have shown limited activity against relapsed medulloblastoma. Temozolomide might serve as backbone for new combinations. Novel and more homogenous trial designs might facilitate the development of new drugs.
Collapse
Affiliation(s)
- Francisco Bautista
- CNIO‐HNJ Clinical Research UnitPediatric Oncology, Hematology and Stem Cell Transplant DepartmentHospital Infantil Universitario Niño JesúsAvenida Menéndez Pelayo, 6528009MadridSpain
| | - Victoria Fioravantti
- CNIO‐HNJ Clinical Research UnitPediatric Oncology, Hematology and Stem Cell Transplant DepartmentHospital Infantil Universitario Niño JesúsAvenida Menéndez Pelayo, 6528009MadridSpain
| | - Teresa de Rojas
- CNIO‐HNJ Clinical Research UnitPediatric Oncology, Hematology and Stem Cell Transplant DepartmentHospital Infantil Universitario Niño JesúsAvenida Menéndez Pelayo, 6528009MadridSpain
| | - Fernando Carceller
- Pediatric and Adolescent Drug Development, Children and Young People's UnitThe Royal Marsden NHS Foundation TrustLondonUK
- Division of Clinical Studies and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Luis Madero
- CNIO‐HNJ Clinical Research UnitPediatric Oncology, Hematology and Stem Cell Transplant DepartmentHospital Infantil Universitario Niño JesúsAvenida Menéndez Pelayo, 6528009MadridSpain
| | - Alvaro Lassaletta
- CNIO‐HNJ Clinical Research UnitPediatric Oncology, Hematology and Stem Cell Transplant DepartmentHospital Infantil Universitario Niño JesúsAvenida Menéndez Pelayo, 6528009MadridSpain
| | - Lucas Moreno
- CNIO‐HNJ Clinical Research UnitPediatric Oncology, Hematology and Stem Cell Transplant DepartmentHospital Infantil Universitario Niño JesúsAvenida Menéndez Pelayo, 6528009MadridSpain
- Instituto de Investigación La PrincesaMadridSpain
| |
Collapse
|
18
|
Steppan DA, Pratilas CA, Loeb DM. Targeted therapy for soft tissue sarcomas in adolescents and young adults. Adolesc Health Med Ther 2017; 8:41-55. [PMID: 28408855 PMCID: PMC5384699 DOI: 10.2147/ahmt.s70377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Soft tissue sarcomas (STSs) are a heterogeneous group of tumors originating from the mesenchyme. Even though they affect individuals in all age groups, the prevalence of subtypes of STSs changes significantly from childhood through adolescence into adulthood. The mainstay of therapy is surgery, with or without the addition of chemotherapy and/or radiation therapy. These treatment modalities are associated, in many cases, with significant morbidity and, given the heterogeneity of tumor histologies encompassed by the term "STS", have not uniformly improved outcomes. Moreover, some subgroups of STSs appear to be more, and others less, responsive to conventional chemotherapy agents. Over the last two decades, our understanding of the biology of STSs is slowly increasing, allowing for the development of more targeted therapies. We review the new treatment modalities that have been tested on patients with STSs, with a special focus on adolescents and young adults, a group of patients that is often underrepresented in clinical trials and has not received the dedicated attention it deserves, given the significant differences in biology and treatment response in comparison to children and adults.
Collapse
Affiliation(s)
- Diana A Steppan
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christine A Pratilas
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David M Loeb
- Division of Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Vo KT, Matthay KK, DuBois SG. Targeted antiangiogenic agents in combination with cytotoxic chemotherapy in preclinical and clinical studies in sarcoma. Clin Sarcoma Res 2016; 6:9. [PMID: 27274393 PMCID: PMC4896001 DOI: 10.1186/s13569-016-0049-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023] Open
Abstract
Sarcomas are a heterogeneous group of mesenchymal malignancies. In recent years, studies have demonstrated that inhibition of angiogenic pathways or disruption of established vasculature can attenuate the growth of sarcomas. However, when used as monotherapy in the clinical setting, these targeted antiangiogenic agents have only provided modest survival benefits in some sarcoma subtypes, and have not been efficacious in others. Preclinical and early clinical data suggest that the addition of conventional chemotherapy to antiangiogenic agents may lead to more effective therapies for patients with these tumors. In the current review, the authors summarize the available evidence and possible mechanisms supporting this approach.
Collapse
Affiliation(s)
- Kieuhoa T. Vo
- />Department of Pediatrics, UCSF School of Medicine, San Francisco School of Medicine, UCSF Benioff Children’s Hospital, University of California, 550 16th Street, 4th Floor, Box 0434, San Francisco, CA 94158 USA
| | - Katherine K. Matthay
- />Department of Pediatrics, UCSF School of Medicine, San Francisco School of Medicine, UCSF Benioff Children’s Hospital, University of California, 550 16th Street, 4th Floor, Box 0434, San Francisco, CA 94158 USA
| | - Steven G. DuBois
- />Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, 450 Brookline Avenue, Dana 3, Boston, MA 02215 USA
| |
Collapse
|
20
|
Bonney PA, Santucci JA, Maurer AJ, Sughrue ME, McNall-Knapp RY, Battiste JD. Dramatic response to temozolomide, irinotecan, and bevacizumab for recurrent medulloblastoma with widespread osseous metastases. J Clin Neurosci 2016; 26:161-3. [DOI: 10.1016/j.jocn.2015.10.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/25/2015] [Indexed: 01/23/2023]
|
21
|
Wagner LM. Fifteen years of irinotecan therapy for pediatric sarcoma: where to next? Clin Sarcoma Res 2015; 5:20. [PMID: 26322224 PMCID: PMC4552408 DOI: 10.1186/s13569-015-0035-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022] Open
Abstract
Over the past 15 years, irinotecan has emerged as an important agent for treating pediatric sarcoma patients. This review summarizes the activity noted in previous studies, and outlines current issues regarding scheduling, route of administration, and amelioration of side effects. Also discussed are new pegylated and nanoliposomal formulations of irinotecan and its active metabolite, SN-38, as well as future plans for how irinotecan may be used in combination with other conventional cytotoxic as well as targeted agents.
Collapse
Affiliation(s)
- Lars M Wagner
- Division of Pediatric Hematology/Oncology, Kentucky Clinic Suite, University of Kentucky, J-457, Lexington, KY 40536 USA
| |
Collapse
|
22
|
Roberts SS, Chou AJ, Cheung NKV. Immunotherapy of Childhood Sarcomas. Front Oncol 2015; 5:181. [PMID: 26301204 PMCID: PMC4528283 DOI: 10.3389/fonc.2015.00181] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/23/2015] [Indexed: 12/29/2022] Open
Abstract
Pediatric sarcomas are a heterogeneous group of malignant tumors of bone and soft tissue origin. Although more than 100 different histologic subtypes have been described, the majority of pediatric cases belong to the Ewing’s family of tumors, rhabdomyosarcoma and osteosarcoma. Most patients that present with localized stage are curable with surgery and/or chemotherapy; however, those with metastatic disease at diagnosis or those who experience a relapse continue to have a very poor prognosis. New therapies for these patients are urgently needed. Immunotherapy is an established treatment modality for both liquid and solid tumors, and in pediatrics, most notably for neuroblastoma and osteosarcoma. In the past, immunomodulatory agents such as interferon, interleukin-2, and liposomal-muramyl tripeptide phosphatidyl-ethanolamine have been tried, with some activity seen in subsets of patients; additionally, various cancer vaccines have been studied with possible benefit. Monoclonal antibody therapies against tumor antigens such as disialoganglioside GD2 or immune checkpoint targets such as CTLA-4 and PD-1 are being actively explored in pediatric sarcomas. Building on the success of adoptive T cell therapy for EBV-related lymphoma, strategies to redirect T cells using chimeric antigen receptors and bispecific antibodies are rapidly evolving with potential for the treatment of sarcomas. This review will focus on recent preclinical and clinical developments in targeted agents for pediatric sarcomas with emphasis on the immunobiology of immune checkpoints, immunoediting, tumor microenvironment, antibody engineering, cell engineering, and tumor vaccines. The future integration of antibody-based and cell-based therapies into an overall treatment strategy of sarcoma will be discussed.
Collapse
Affiliation(s)
- Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| | - Alexander J Chou
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center , New York, NY , USA
| |
Collapse
|
23
|
Buczkowicz P, Hawkins C. Pathology, Molecular Genetics, and Epigenetics of Diffuse Intrinsic Pontine Glioma. Front Oncol 2015; 5:147. [PMID: 26175967 PMCID: PMC4485076 DOI: 10.3389/fonc.2015.00147] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/16/2015] [Indexed: 11/13/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a devastating pediatric brain cancer with no effective therapy. Histological similarity of DIPG to supratentorial high-grade astrocytomas of adults has led to assumptions that these entities possess similar underlying molecular properties and therefore similar therapeutic responses to standard therapies. The failure of all clinical trials in the last 30 years to improve DIPG patient outcome has suggested otherwise. Recent studies employing next-generation sequencing and microarray technologies have provided a breadth of evidence highlighting the unique molecular genetics and epigenetics of this cancer, distinguishing it from both adult and pediatric cerebral high-grade astrocytomas. This review describes the most common molecular genetic and epigenetic signatures of DIPG in the context of molecular subgroups and histopathological diagnosis, including this tumor entity's unique mutational landscape, copy number alterations, and structural variants, as well as epigenetic changes on the global DNA and histone levels. The increased knowledge of DIPG biology and histopathology has opened doors to new diagnostic and therapeutic avenues.
Collapse
Affiliation(s)
- Pawel Buczkowicz
- Division of Pathology, The Hospital for Sick Children , Toronto, ON , Canada ; The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children , Toronto, ON , Canada
| | - Cynthia Hawkins
- Division of Pathology, The Hospital for Sick Children , Toronto, ON , Canada ; The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children , Toronto, ON , Canada ; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
24
|
Capitini CM, Otto M, DeSantes KB, Sondel PM. Immunotherapy in pediatric malignancies: current status and future perspectives. Future Oncol 2015; 10:1659-78. [PMID: 25145434 DOI: 10.2217/fon.14.62] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Novel immune-based therapies are becoming available as additions to, and in some cases as alternatives to, the traditional treatment modalities such as chemotherapy, surgery and radiation that have improved outcomes for childhood cancer for decades. In this article, we will discuss what immunotherapies are being tested in the clinic, barriers to widespread application, and the future of immuno-oncology for childhood cancer. While in many cases, these therapies have shown dramatic responses in the setting of refractory or relapsed cancer, much remains to be learned about how to integrate these therapies into existing upfront regimens. The progress and challenges of developing immunotherapies for childhood cancer in a timely and cost-effective fashion will be discussed.
Collapse
Affiliation(s)
- Christian M Capitini
- Department of Pediatrics & Carbone Cancer Center, University of Wisconsin School of Medicine & Public Health, 1111 Highland Avenue, WIMR 4137, Madison, WI 53705, USA
| | | | | | | |
Collapse
|
25
|
A. Shah P, Department of Pediatrics, Division of Hematology/Oncology, Jackson Memorial Hospital, University of Miami, Miami, FL 33136, USA, Goldberg J. Novel Approaches to Pediatric Cancer: Immunotherapy. AIMS MEDICAL SCIENCE 2015. [DOI: 10.3934/medsci.2015.2.104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
26
|
Abstract
Four out of five children diagnosed with cancer can be cured with contemporary cancer therapy. This represents a dramatic improvement since 50 years ago when the cure rate of childhood cancer was <25% in the pre-chemotherapy era. Over the past ten years, while improvement in overall survival (OS) has been marginal, progress in pediatric oncology lies with adopting risk-adapted therapeutic approach. This has been made possible through identifying clinical and biologic prognostic factors with rigorous research and stratifying patients using these risk factors, and subsequently modifying therapy according to risk group assignment. This review provides a perspective for eight distinct pediatric malignancies, in which significant advances in treatment were made in the last decade and are leading to changes in standard of care. This includes four hematologic malignancies [acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), non-Hodgkin lymphoma (NHL) and Hodgkin lymphoma (HL)] and four solid tumors [medulloblastoma (MB), low grade glioma (LGG), neuroblastoma (NB) and Ewing sarcoma (ES)]. Together, they comprise 60% of childhood cancer. Improved patient outcome is not limited to better survival, but encompasses reducing both short and long-term treatment-related complications which is as important as cure, given the majority of childhood cancer patients will become long-term survivors. Risk-adapted approach allows treatment intensification in the high-risk cohort while therapy can be de-escalated in the low-risk to minimize toxicity and late sequelae without compromising survival. Advances in medical research technology have also led to a rapid increase in the understanding of the genetics of childhood cancer in the last decade, facilitating identification of molecular targets that can potentially be exploited for therapeutic benefits. As we move into the era of targeted therapeutics, searching for novel agents that target specific genetic lesions becomes a major research focus. We provide an overview of seven novel agents (bevacizumab, bortezomib, vorinostat, sorafenib, tipifarnib, erlotinib and mTOR inhibitors), which have been most frequently pursued in childhood cancers in the last decade, as well as reporting the progress of clinical trials involving these agents.
Collapse
Affiliation(s)
- Federica Saletta
- 1 Children's Cancer Research Unit, Kid's Research Institute, The Children's Hospital at Westmead, Westmead, NSW, Australia ; 2 Oncology Department, The Children's Hospital at Westmead, Westmead, NSW, Australia ; 3 Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Michaela S Seng
- 1 Children's Cancer Research Unit, Kid's Research Institute, The Children's Hospital at Westmead, Westmead, NSW, Australia ; 2 Oncology Department, The Children's Hospital at Westmead, Westmead, NSW, Australia ; 3 Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Loretta M S Lau
- 1 Children's Cancer Research Unit, Kid's Research Institute, The Children's Hospital at Westmead, Westmead, NSW, Australia ; 2 Oncology Department, The Children's Hospital at Westmead, Westmead, NSW, Australia ; 3 Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|