1
|
Karam S, Gebreil A, Alksas A, Balaha HM, Khalil A, Ghazal M, Contractor S, El-Baz A. Insights into Personalized Care Strategies for Wilms Tumor: A Narrative Literature Review. Biomedicines 2024; 12:1455. [PMID: 39062028 PMCID: PMC11274555 DOI: 10.3390/biomedicines12071455] [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: 05/02/2024] [Revised: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Wilms tumor (WT), or nephroblastoma, is the predominant renal malignancy in the pediatric population. This narrative review explores the evolution of personalized care strategies for WT, synthesizing critical developments in molecular diagnostics and treatment approaches to enhance patient-specific outcomes. We surveyed recent literature from the last five years, focusing on high-impact research across major databases such as PubMed, Scopus, and Web of Science. Diagnostic advancements, including liquid biopsies and diffusion-weighted MRI, have improved early detection precision. The prognostic significance of genetic markers, particularly WT1 mutations and miRNA profiles, is discussed. Novel predictive tools integrating genetic and clinical data to anticipate disease trajectory and therapy response are explored. Progressive treatment strategies, particularly immunotherapy and targeted agents such as HIF-2α inhibitors and GD2-targeted immunotherapy, are highlighted for their role in personalized treatment protocols, especially for refractory or recurrent WT. This review underscores the necessity for personalized management supported by genetic insights, with improved survival rates for localized disease exceeding 90%. However, knowledge gaps persist in therapies for high-risk patients and strategies to reduce long-term treatment-related morbidity. In conclusion, this narrative review highlights the need for ongoing research, particularly on the long-term outcomes of emerging therapies and integrating multi-omic data to inform clinical decision-making, paving the way for more individualized treatment pathways.
Collapse
Affiliation(s)
- Salma Karam
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (S.K.); (A.G.); (A.A.); (H.M.B.)
| | - Ahmad Gebreil
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (S.K.); (A.G.); (A.A.); (H.M.B.)
| | - Ahmed Alksas
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (S.K.); (A.G.); (A.A.); (H.M.B.)
| | - Hossam Magdy Balaha
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (S.K.); (A.G.); (A.A.); (H.M.B.)
| | - Ashraf Khalil
- College of Technological Innovation, Zayed University, Abu Dhabi 4783, United Arab Emirates;
| | - Mohammed Ghazal
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates;
| | - Sohail Contractor
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA;
| | - Ayman El-Baz
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA; (S.K.); (A.G.); (A.A.); (H.M.B.)
| |
Collapse
|
2
|
Du S, Zhao Y, Hu Z, Liu S, Roy A, Shen J, Zhu L, Hamuro L. Pediatric model-based dose optimization using a pooled exposure-response safety analysis for nivolumab and nivolumab plus ipilimumab combination in melanoma. CPT Pharmacometrics Syst Pharmacol 2024; 13:168-179. [PMID: 37873561 PMCID: PMC10787196 DOI: 10.1002/psp4.13070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023] Open
Abstract
An exposure-response (E-R) safety analysis was conducted across adult and pediatric (<18 years) studies to evaluate the potential impact of higher nivolumab and/or ipilimumab exposures in adolescents (≥12 to <18 years) versus adults with melanoma using the approved adult dosing regimens for nivolumab alone or in combination with ipilimumab. Data from 3507 patients across 15 studies were used to examine the relationship between nivolumab-ipilimumab daily average exposure and time to grade 2+ immune-mediated adverse events (gr2+ IMAEs). Results from the E-R safety model showed ipilimumab, but not nivolumab, exposure to be a statistically significant predictor of gr2+ IMAEs. Significant covariates included sex (41% higher risk for women than men), line of therapy (19% higher for first-line than later-line), and treatment setting (26% lower for adjuvant than advanced melanoma). Younger age and lower body weight (BW) were each associated with a lower risk of gr2+ IMAEs (hazard ratio [HR]: 0.830 for 15-year-olds versus 60-year-olds and 0.84 for BW 52 kg versus 75 kg). For adolescents with melanoma treated with nivolumab in the advanced or adjuvant settings, these results are supportive of nivolumab flat dosing regimens for adolescents greater than or equal to 40 kg and BW-based dosing for adolescents less than 40 kg. These results also support adult weight-based dosing regimens for nivolumab plus ipilimumab in adolescents with advanced melanoma. This analysis suggests that although higher exposures are predicted in adolescents with lower weight compared with adults, there is no predicted immune-mediated safety risk when treated with the approved adult dosing of nivolumab with/without ipilimumab.
Collapse
Affiliation(s)
| | - Yue Zhao
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Zheyi Hu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Sihang Liu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Jun Shen
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Li Zhu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | |
Collapse
|
3
|
Rohila D, Park IH, Pham TV, Jones R, Tapia E, Liu KX, Tamayo P, Yu A, Sharabi AB, Joshi S. Targeting macrophage Syk enhances responses to immune checkpoint blockade and radiotherapy in high-risk neuroblastoma. Front Immunol 2023; 14:1148317. [PMID: 37350973 PMCID: PMC10283071 DOI: 10.3389/fimmu.2023.1148317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/02/2023] [Indexed: 06/24/2023] Open
Abstract
Background Neuroblastoma (NB) is considered an immunologically cold tumor and is usually less responsive to immune checkpoint blockade (ICB). Tumor-associated macrophages (TAMs) are highly infiltrated in NB tumors and promote immune escape and resistance to ICB. Hence therapeutic strategies targeting immunosuppressive TAMs can improve responses to ICB in NB. We recently discovered that spleen tyrosine kinase (Syk) reprograms TAMs toward an immunostimulatory phenotype and enhances T-cell responses in the lung adenocarcinoma model. Here we investigated if Syk is an immune-oncology target in NB and tested whether a novel immunotherapeutic approach utilizing Syk inhibitor together with radiation and ICB could provide a durable anti-tumor immune response in an MYCN amplified murine model of NB. Methods Myeloid Syk KO mice and syngeneic MYCN-amplified cell lines were used to elucidate the effect of myeloid Syk on the NB tumor microenvironment (TME). In addition, the effect of Syk inhibitor, R788, on anti-tumor immunity alone or in combination with anti-PDL1 mAb and radiation was also determined in murine NB models. The underlying mechanism of action of this novel therapeutic combination was also investigated. Results Herein, we report that Syk is a marker of NB-associated macrophages and plays a crucial role in promoting immunosuppression in the NB TME. We found that the blockade of Syk in NB-bearing mice markedly impairs tumor growth. This effect is facilitated by macrophages that become immunogenic in the absence of Syk, skewing the suppressive TME towards immunostimulation and activating anti-tumor immune responses. Moreover, combining FDA-approved Syk inhibitor, R788 (fostamatinib) along with anti-PDL1 mAb provides a synergistic effect leading to complete tumor regression and durable anti-tumor immunity in mice bearing small tumors (50 mm3) but not larger tumors (250 mm3). However, combining radiation to R788 and anti-PDL1 mAb prolongs the survival of mice bearing large NB9464 tumors. Conclusion Collectively, our findings demonstrate the central role of macrophage Syk in NB progression and demonstrate that Syk blockade can "reeducate" TAMs towards immunostimulatory phenotype, leading to enhanced T cell responses. These findings further support the clinical evaluation of fostamatinib alone or with radiation and ICB, as a novel therapeutic intervention in neuroblastoma.
Collapse
Affiliation(s)
- Deepak Rohila
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - In Hwan Park
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Timothy V. Pham
- Office of Cancer Genomics, University of California San Diego, San Diego, CA, United States
| | - Riley Jones
- Department of Radiation Medicine and Applied Sciences, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Elisabette Tapia
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Kevin X. Liu
- Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, MA, United States
| | - Pablo Tamayo
- Office of Cancer Genomics, University of California San Diego, San Diego, CA, United States
| | - Alice Yu
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Andrew B. Sharabi
- Department of Radiation Medicine and Applied Sciences, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Shweta Joshi
- Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
4
|
Yu EY, Cheung NKV, Lue NF. Connecting telomere maintenance and regulation to the developmental origin and differentiation states of neuroblastoma tumor cells. J Hematol Oncol 2022; 15:117. [PMID: 36030273 PMCID: PMC9420296 DOI: 10.1186/s13045-022-01337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature that distinguishes clinically high-risk neuroblastoma from low-risk tumors is telomere maintenance. Specifically, neuroblastoma tumors with either active telomerase or alternative lengthening of telomeres exhibit aggressive growth characteristics that lead to poor outcomes, whereas tumors without telomere maintenance can be managed with observation or minimal treatment. Even though the need for cancer cells to maintain telomere DNA-in order to sustain cell proliferation-is well established, recent studies suggest that the neural crest origin of neuroblastoma may enforce unique relationships between telomeres and tumor malignancy. Specifically in neuroblastoma, telomere structure and telomerase activity are correlated with the adrenergic/mesenchymal differentiation states, and manipulating telomerase activity can trigger tumor cell differentiation. Both findings may reflect features of normal neural crest development. This review summarizes recent advances in the characterization of telomere structure and telomere maintenance mechanisms in neuroblastoma and discusses the findings in the context of relevant literature on telomeres during embryonic and neural development. Understanding the canonical and non-canonical roles of telomere maintenance in neuroblastoma could reveal vulnerabilities for telomere-directed therapies with potential applications to other pediatric malignancies.
Collapse
Affiliation(s)
- Eun Young Yu
- Department of Microbiology & Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Neal F Lue
- Department of Microbiology & Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
5
|
Nguyen T, Mueller S, Malbari F. Review: Neurological Complications From Therapies for Pediatric Brain Tumors. Front Oncol 2022; 12:853034. [PMID: 35480100 PMCID: PMC9035987 DOI: 10.3389/fonc.2022.853034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022] Open
Abstract
Surgery, chemotherapy and radiation have been the mainstay of pediatric brain tumor treatment over the past decades. Recently, new treatment modalities have emerged for the management of pediatric brain tumors. These therapies range from novel radiotherapy techniques and targeted immunotherapies to checkpoint inhibitors and T cell transfer therapies. These treatments are currently investigated with the goal of improving survival and decreasing morbidity. However, compared to traditional therapies, these novel modalities are not as well elucidated and similarly has the potential to cause significant short and long-term sequelae, impacting quality of life. Treatment complications are commonly mediated through direct drug toxicity or vascular, infectious, or autoimmune mechanisms, ranging from immune effector cell associated neurotoxicity syndrome with CART-cells to neuropathy with checkpoint inhibitors. Addressing treatment-induced complications is the focus of new trials, specifically improving neurocognitive outcomes. The aim of this review is to explore the pathophysiology underlying treatment related neurologic side effects, highlight associated complications, and describe the future direction of brain tumor protocols. Increasing awareness of these neurologic complications from novel therapies underscores the need for quality-of-life metrics and considerations in clinical trials to decrease associated treatment-induced morbidity.
Collapse
Affiliation(s)
- Thien Nguyen
- Department of Pediatrics, University of San Francisco, San Francisco, CA, United States
- *Correspondence: Thien Nguyen,
| | - Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of San Francisco, San Francisco, CA, United States
| | - Fatema Malbari
- Division of Neurology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
6
|
Dasgupta A, Tsay E, Federman N, Lechner MG, Su MA. Polyendocrine Autoimmunity and Diabetic Ketoacidosis Following Anti-PD-1 and Interferon α. Pediatrics 2022; 149:e2021053363. [PMID: 35274131 PMCID: PMC9804498 DOI: 10.1542/peds.2021-053363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 01/03/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) therapies are now first-line therapy for many advanced malignancies in adults, with emerging use in children. With increasing ICI use, prompt recognition and optimal management of ICI-associated immune-related adverse events (IRAEs) are critical. Nearly 60% of ICI-treated adults develop IRAEs, which commonly manifest as autoimmune skin, gastrointestinal, and endocrine disease and can be life-threatening. The incidence, presentation, and disease course of spontaneous autoimmune diseases differ between adults and children, but the pattern of pediatric IRAEs is currently unclear. We report a case of a pediatric patient presenting with new onset autoimmune diabetes mellitus and diabetic ketoacidosis during ICI treatment of fibrolamellar hepatocellular carcinoma (FLC). Distinct from spontaneous type 1 diabetes mellitus (T1DM), this patient progressed rapidly and was negative for known β cell autoantibodies. Additionally, the patient was positive for 21-hydroxylase autoantibodies, suggesting development of concomitant adrenal autoimmunity. Current guidelines for the management of IRAEs in adults may not be appropriate for the management of pediatric patients, who may have different autoimmune risks in a developmental context.
Collapse
Affiliation(s)
- Aditi Dasgupta
- Dept. of Pediatrics, UCLA Geffen School of Medicine, Los Angeles, CA
| | - Eric Tsay
- Div. of Pediatric Endocrinology, UCLA Geffen School of Medicine, Los Angeles, CA
| | - Noah Federman
- Div. of Pediatric Hematology and Oncology, UCLA Geffen School of Medicine, Los Angeles, CA
| | - Melissa G. Lechner
- Div. of Endocrinology, Diabetes, and Metabolism, UCLA Geffen School of Medicine, Los Angeles, CA
| | - Maureen A. Su
- Div. of Pediatric Endocrinology, UCLA Geffen School of Medicine, Los Angeles, CA
- Dept. of Microbiology, Immunology, and Molecular Genetics, UCLA Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|
7
|
The New Treatment Methods for Non-Hodgkin Lymphoma in Pediatric Patients. Cancers (Basel) 2022; 14:cancers14061569. [PMID: 35326719 PMCID: PMC8945992 DOI: 10.3390/cancers14061569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
One of the most common cancer malignancies is non-Hodgkin lymphoma, whose incidence is nearly 3% of all 36 cancers combined. It is the fourth highest cancer occurrence in children and accounts for 7% of cancers in patients under 20 years of age. Today, the survivability of individuals diagnosed with non-Hodgkin lymphoma varies by about 70%. Chemotherapy, radiation, stem cell transplantation, and immunotherapy have been the main methods of treatment, which have improved outcomes for many oncological patients. However, there is still the need for creation of novel medications for those who are treatment resistant. Additionally, more effective drugs are necessary. This review gathers the latest findings on non-Hodgkin lymphoma treatment options for pediatric patients. Attention will be focused on the most prominent therapies such as monoclonal antibodies, antibody–drug conjugates, chimeric antigen receptor T cell therapy and others.
Collapse
|
8
|
Anti-GD2 Directed Immunotherapy for High-Risk and Metastatic Neuroblastoma. Biomolecules 2022; 12:biom12030358. [PMID: 35327550 PMCID: PMC8945428 DOI: 10.3390/biom12030358] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroblastoma is one of the few childhood cancers that carries a tumor-specific antigen in the form of a glycolipid antigen known as GD2. It has restricted expression in normal tissue, such as peripheral afferent nerves. Monoclonal antibodies targeting GD2 have been applied clinically to high-risk neuroblastoma with significant success. However, there are different anti-GD2 products and administration regimens. For example, anti-GD2 has been used in combination with chemotherapy during the induction phase or with retinoic acid during the maintenance stage. Regimens also vary in the choice of whether to add cytokines (i.e., IL-2, GMCSF, or both). Furthermore, the addition of an immune enhancer, such as β-glucan, or allogeneic natural killer cells also becomes a confounder in the interpretation. The question concerning which product or method of administration is superior remains to be determined. So far, most studies agree that adding anti-GD2 to the conventional treatment protocol can achieve better short- to intermediate-term event-free and overall survival, but the long-term efficacy remains to be verified. How to improve its efficacy is another challenge. Late relapse and central nervous system metastasis have emerged as new problems. The methods to overcome the mechanisms related to immune evasion or resistance to immunotherapy represent new challenges to be resolved. The newer anti-GD2 strategies, such as bispecific antibody linking of anti-GD2 with activated T cells or chimeric antigen receptor T cells, are currently under clinical trials, and they may become promising alternatives. The use of anti-GD2/GD3 tumor vaccine is a novel and potential approach to minimizing late relapse. How to induce GD2 expression from tumor cells using the epigenetic approach is a hot topic nowadays. We expect that anti-GD2 treatment can serve as a model for the use of monoclonal antibody immunotherapy against cancers in the future.
Collapse
|
9
|
Expression of Immunomodulatory Checkpoint Molecules in Drug-Resistant Neuroblastoma: An Exploratory Study. Cancers (Basel) 2022; 14:cancers14030751. [PMID: 35159017 PMCID: PMC8833944 DOI: 10.3390/cancers14030751] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Neuroblastoma is a common childhood cancer with poor prognosis. Prior studies suggest that inhibition of molecules called checkpoint proteins, which normally prevent one’s own immune system from attacking itself, has been successfully used for treatment of multiple advanced adult cancers but has yet to be fully explored in neuroblastoma. Cancer can hijack these pathways to prevent the immune system from recognizing and destroying cancer cells. We investigated checkpoint protein expression in pediatric neuroblastoma and its role in drug resistance. We created drug-resistant neuroblastoma cell lines and compared expression of checkpoint proteins between drug-resistant and parental cell lines. In total, 13 checkpoint proteins were expressed by all cell lines regardless of drug resistance. Although PD-L1 and checkpoint proteins do not necessarily impart drug resistance, they may be potential targets for drug therapy. Benchmarking checkpoint proteins provides the basis for future studies identifying targets for directed therapy and biomarkers for cancer detection or prognosis. Abstract Neuroblastoma is a common childhood cancer with poor prognosis when at its advanced stage. Checkpoint molecule inhibition is successful in treating multiple advanced adult cancers. We investigated PD-L1 and other checkpoint molecule expression to determine their roles in drug resistance and usefulness as targets for drug therapy. We developed three doxorubicin-resistant (DoxR) cell lines from parental cell lines. Matrigel in vitro invasion assays were used to compare invasiveness. Western blot assays were used to compare PD-L1 expression. Immuno-oncology checkpoint protein panels were used to compare concentrations of 17 checkpoint molecules both cellular and soluble. PD-L1 and 12 other checkpoint molecules were present in all cell lysates of each cell line without significantly different levels. Three were solubilized in the media of each cell line. PD-L1 is expressed in all DoxR and parental neuroblastoma cells and may be a potential target for drug therapy although its role in drug resistance remains unclear. Benchmarking checkpoint molecules provides the basis for future studies identifying targets for directed therapy and biomarkers for cancer detection or prognosis.
Collapse
|
10
|
Brickler M, Raskin A, Ryan TD. Current State of Pediatric Cardio-Oncology: A Review. CHILDREN (BASEL, SWITZERLAND) 2022; 9:127. [PMID: 35204848 PMCID: PMC8870613 DOI: 10.3390/children9020127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 01/03/2023]
Abstract
The landscape of pediatric oncology has dramatically changed over the course of the past several decades with five-year survival rates surpassing 80%. Anthracycline therapy has been the cornerstone of many chemotherapy regimens for pediatric patients since its introduction in the 1960s, and recent improved survival has been in large part due to advancements in chemotherapy, refinement of supportive care treatments, and development of novel therapeutics such as small molecule inhibitors, chimeric antigen receptor T-cell therapy, and immune checkpoint inhibitors. Unfortunately, many cancer-targeted therapies can lead to acute and chronic cardiovascular pathologies. The range of cardiotoxicity can vary but includes symptomatic or asymptotic heart failure, arrhythmias, coronary artery disease, valvar disease, pericardial disease, hypertension, and peripheral vascular disease. There is lack of data guiding primary prevention and treatment strategies in the pediatric population, which leads to substantial practice variability. Several important future research directions have been identified, including as they relate to cardiac disease, prevention strategies, management of cardiovascular risk factors, risk prediction, early detection, and the role of genetic susceptibility in development of cardiotoxicity. Continued collaborative research will be key in advancing the field. The ideal model for pediatric cardio-oncology is a proactive partnership between pediatric cardiologists and oncologists in order to better understand, treat, and ideally prevent cardiac disease in pediatric oncology patients.
Collapse
Affiliation(s)
| | | | - Thomas D. Ryan
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| |
Collapse
|
11
|
Pediatric glioblastoma: mechanisms of immune evasion and potential therapeutic opportunities. Cancer Immunol Immunother 2022; 71:1813-1822. [PMID: 35020009 DOI: 10.1007/s00262-021-03131-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
Pediatric glioblastoma is relatively rare compared with its adult counterpart but is associated with a similarly grim prognosis. Available data indicate that pediatric glioblastomas are molecularly distinct from adult tumors, and relatively little is known about the pediatric glioblastoma tumor microenvironment (TME). Cancer immunotherapy has emerged as a new pillar of cancer treatment and is revolutionizing the care of patients with many advanced solid tumors, including melanoma, non-small cell lung cancer, head and neck cancer, and renal cell carcinoma. Unfortunately, attempts to treat adult glioblastoma with current immunotherapies have had limited success to date. Nevertheless, the immune milieu in pediatric glioblastoma is distinct from that found in adult tumors, and evidence suggests that pediatric tumors are less immunosuppressive. As a result, immunotherapies should be specifically evaluated in the pediatric context. The purpose of this review is to explore known and emerging mechanisms of immune evasion in pediatric glioblastoma and highlight potential opportunities for implementing immunotherapy in the treatment of these devastating pediatric brain tumors.
Collapse
|
12
|
Gonçalves FG, Viaene AN, Vossough A. Advanced Magnetic Resonance Imaging in Pediatric Glioblastomas. Front Neurol 2021; 12:733323. [PMID: 34858308 PMCID: PMC8631300 DOI: 10.3389/fneur.2021.733323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/12/2021] [Indexed: 12/26/2022] Open
Abstract
The shortly upcoming 5th edition of the World Health Organization Classification of Tumors of the Central Nervous System is bringing extensive changes in the terminology of diffuse high-grade gliomas (DHGGs). Previously "glioblastoma," as a descriptive entity, could have been applied to classify some tumors from the family of pediatric or adult DHGGs. However, now the term "glioblastoma" has been divested and is no longer applied to tumors in the family of pediatric types of DHGGs. As an entity, glioblastoma remains, however, in the family of adult types of diffuse gliomas under the insignia of "glioblastoma, IDH-wildtype." Of note, glioblastomas still can be detected in children when glioblastoma, IDH-wildtype is found in this population, despite being much more common in adults. Despite the separation from the family of pediatric types of DHGGs, what was previously labeled as "pediatric glioblastomas" still remains with novel labels and as new entities. As a result of advances in molecular biology, most of the previously called "pediatric glioblastomas" are now classified in one of the four family members of pediatric types of DHGGs. In this review, the term glioblastoma is still apocryphally employed mainly due to its historical relevance and the paucity of recent literature dealing with the recently described new entities. Therefore, "glioblastoma" is used here as an umbrella term in the attempt to encompass multiple entities such as astrocytoma, IDH-mutant (grade 4); glioblastoma, IDH-wildtype; diffuse hemispheric glioma, H3 G34-mutant; diffuse pediatric-type high-grade glioma, H3-wildtype and IDH-wildtype; and high grade infant-type hemispheric glioma. Glioblastomas are highly aggressive neoplasms. They may arise anywhere in the developing central nervous system, including the spinal cord. Signs and symptoms are non-specific, typically of short duration, and usually derived from increased intracranial pressure or seizure. Localized symptoms may also occur. The standard of care of "pediatric glioblastomas" is not well-established, typically composed of surgery with maximal safe tumor resection. Subsequent chemoradiation is recommended if the patient is older than 3 years. If younger than 3 years, surgery is followed by chemotherapy. In general, "pediatric glioblastomas" also have a poor prognosis despite surgery and adjuvant therapy. Magnetic resonance imaging (MRI) is the imaging modality of choice for the evaluation of glioblastomas. In addition to the typical conventional MRI features, i.e., highly heterogeneous invasive masses with indistinct borders, mass effect on surrounding structures, and a variable degree of enhancement, the lesions may show restricted diffusion in the solid components, hemorrhage, and increased perfusion, reflecting increased vascularity and angiogenesis. In addition, magnetic resonance spectroscopy has proven helpful in pre- and postsurgical evaluation. Lastly, we will refer to new MRI techniques, which have already been applied in evaluating adult glioblastomas, with promising results, yet not widely utilized in children.
Collapse
Affiliation(s)
- Fabrício Guimarães Gonçalves
- Division of Neuroradiology, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Arastoo Vossough
- Division of Neuroradiology, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
13
|
Que Y, Hu Y, Hong D, Zhang Y. Trends in clinical development of pediatric cancer for PD-1 and PD-L1 inhibitors: an analysis of ClinicalTrials.gov. J Immunother Cancer 2021; 9:jitc-2021-002920. [PMID: 34583971 PMCID: PMC8479973 DOI: 10.1136/jitc-2021-002920] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2021] [Indexed: 01/25/2023] Open
Abstract
Compared with cytotoxic chemotherapy, radiotherapy, and surgery, positive findings have been acquired through the approach of blocking the programmed cell death protein 1 (PD-1) pathway with antibodies that exert inhibitory effects on PD-1 or cell death protein ligand 1 (PD-L1). Results from clinical trials showed great potential in adult patients with cancers, such as melanoma, non-small cell carcinoma, and nasopharyngeal carcinoma. However, studies of checkpoint inhibitors specifically targeting PD-1/PD-L1 in pediatric patients are limited. We evaluated ongoing clinical trials using PD-1 or PD-L1 inhibitors alone or in combination with other therapies to treat pediatric cancer. The proportion of PD-1/PD-L1 combination clinical trials has increased since 2018; the three most common trials over the past 2 years used CTLA-4 monoclonal antibodies, chemotherapy, and therapies that target the vascular endothelial growth factor axis. This commentary aimed to provide trends and specific insights into methods for conducting clinical trials of immunotherapy in the pediatric population.
Collapse
Affiliation(s)
- Yi Que
- Department of Pediatric Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yang Hu
- Department of Pediatric Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Dongchun Hong
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yizhuo Zhang
- Department of Pediatric Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Expression of programmed death ligand 1 in drug-resistant osteosarcoma: An exploratory study. Surg Open Sci 2021; 6:10-14. [PMID: 34386763 PMCID: PMC8346678 DOI: 10.1016/j.sopen.2021.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 01/04/2023] Open
Abstract
Background Inhibition of the programmed death ligand 1, programmed death 1 pathway has been successfully used for treatment of multiple advanced adult cancers. However, its use in pediatric osteosarcoma is still in its infancy. In this study, we investigated programmed death ligand 1 and other checkpoint molecules' expression to determine the potential usefulness as targets for drug therapy. Methods We incubated human wild-type osteosarcoma cells with incremental concentrations of doxorubicin to create a doxorubicin-resistant cell line. Matrigel in vitro invasion assays were used to compare invasiveness. Comparative programmed death ligand 1 expression was evaluated by Western blot assays. An immuno-oncology checkpoint protein panel was used to compare concentrations of 16 other checkpoint molecules. Chi-square tests and Wilcoxon rank-sum tests were used to determine significant differences. Results A doxorubicin-resistant cell line was successfully created and was significantly more invasive than wild-type cells (0.47 vs 0.07, P < .001). On Western blot assay, doxorubicin-resistant but not wild-type cells expressed programmed death ligand 1. Doxorubicin-resistant cells had significantly higher levels of T-cell immunoglobulin-3 and cluster of differentiation 86 and higher cluster of differentiation 27, cluster of differentiation 40, lymphocyte-activation gene-3, cluster of differentiation 80, programmed death ligand 1, programmed death ligand 2, and inducible T-cell costimulatory expression than wild-type cells. Both lines expressed B- and T-lymphocyte attenuator, cluster of differentiation 28, herpesvirus entry mediator, and programmed death 1. Herpesvirus entry mediator, cluster of differentiation 40, and programmed death ligand 2 were also present in the culture media of both cell lines. Conclusion Doxorubicin-resistant osteosarcoma seems to express higher programmed death ligand 1 than nonresistant wild-type cells. Benchmarking checkpoint molecules may provide the basis for future studies that elucidate pathways of drug resistance and tumor metastasis, biomarkers for cancer prognosis or recurrence, and future targets for directed drug therapy.
Collapse
Key Words
- BTLA, B- and T-lymphocyte attenuator
- CD27, cluster of differentiation 27
- CD28, cluster of differentiation 28
- CD40, cluster of differentiation 40
- CD80, cluster of differentiation 80
- CD86, cluster of differentiation 86
- CTLA-4, cytotoxic T-lymphocyte-associated protein 4
- DoxR, doxorubicin resistant
- FDA, Food and Drug Administration
- GITR, glucocorticoid-induced TNFR-related protein
- GITRL, ligand for receptor TNFRSF18/AITR/GITR
- HVEM, herpesvirus entry mediator
- ICOS, inducible T-cell costimulatory (ICOS)
- LAG-3, lymphocyte-activation gene-3
- PD-1, programmed death 1
- PD-L1, programmed death ligand 1
- PD-L2, programmed death ligand 2
- TIM-3, T-cell immunoglobulin-3
- TLR-2, Toll like receptor 2
- WT, wild type
Collapse
|
15
|
Munari E, Mariotti FR, Quatrini L, Bertoglio P, Tumino N, Vacca P, Eccher A, Ciompi F, Brunelli M, Martignoni G, Bogina G, Moretta L. PD-1/PD-L1 in Cancer: Pathophysiological, Diagnostic and Therapeutic Aspects. Int J Mol Sci 2021; 22:5123. [PMID: 34066087 PMCID: PMC8151504 DOI: 10.3390/ijms22105123] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Immune evasion is a key strategy adopted by tumor cells to escape the immune system while promoting their survival and metastatic spreading. Indeed, several mechanisms have been developed by tumors to inhibit immune responses. PD-1 is a cell surface inhibitory receptor, which plays a major physiological role in the maintenance of peripheral tolerance. In pathological conditions, activation of the PD-1/PD-Ls signaling pathway may block immune cell activation, a mechanism exploited by tumor cells to evade the antitumor immune control. Targeting the PD-1/PD-L1 axis has represented a major breakthrough in cancer treatment. Indeed, the success of PD-1 blockade immunotherapies represents an unprecedented success in the treatment of different cancer types. To improve the therapeutic efficacy, a deeper understanding of the mechanisms regulating PD-1 expression and signaling in the tumor context is required. We provide an overview of the current knowledge of PD-1 expression on both tumor-infiltrating T and NK cells, summarizing the recent evidence on the stimuli regulating its expression. We also highlight perspectives and limitations of the role of PD-L1 expression as a predictive marker, discuss well-established and novel potential approaches to improve patient selection and clinical outcome and summarize current indications for anti-PD1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Enrico Munari
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy;
| | - Francesca R. Mariotti
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Linda Quatrini
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Pietro Bertoglio
- Division of Thoracic Surgery, IRCCS Maggiore Teaching Hospital and Sant’Orsola University Hospital, 40133 Bologna, Italy;
| | - Nicola Tumino
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Paola Vacca
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Albino Eccher
- Pathology Unit, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Francesco Ciompi
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, 6543 SH Nijmegen, The Netherlands;
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (M.B.); (G.M.)
| | - Guido Martignoni
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (M.B.); (G.M.)
- Pathology Unit, Pederzoli Hospital, 37019 Peschiera del Garda, Italy
| | - Giuseppe Bogina
- Pathology Unit, IRCCS Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Lorenzo Moretta
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| |
Collapse
|
16
|
Vatner R, James CD, Sathiaseelan V, Bondra KM, Kalapurakal JA, Houghton PJ. Radiation therapy and molecular-targeted agents in preclinical testing for immunotherapy, brain tumors, and sarcomas: Opportunities and challenges. Pediatr Blood Cancer 2021; 68 Suppl 2:e28439. [PMID: 32827353 DOI: 10.1002/pbc.28439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 01/07/2023]
Abstract
Despite radiation therapy (RT) being an integral part of the treatment of most pediatric cancers and the recent discovery of novel molecular-targeted agents (MTAs) in this era of precision medicine with the potential to improve the therapeutic ratio of modern chemoradiotherapy regimens, there are only a few preclinical trials being conducted to discover novel radiosensitizers and radioprotectors. This has resulted in a paucity of translational clinical trials combining RT and novel MTAs. This report describes the opportunities and challenges of investigating RT together with MTAs in preclinical testing for immunotherapy, brain tumors, and sarcomas in pediatric oncology. We discuss the need for improving the collaboration between radiation oncologists, biologists, and physicists to improve the reliability, reproducibility, and translational potential of RT-based preclinical research. Current translational clinical trials using RT and MTAs for immunotherapy, brain tumors, and sarcomas are described. The technologic advances in experimental RT, availability of novel experimental tumor models, advances in immunology and tumor biology, and the discovery of novel MTAs together hold considerable promise for good quality preclinical and clinical multimodality research to improve the current rates of survival and toxicity in children afflicted with cancer.
Collapse
Affiliation(s)
- Ralph Vatner
- Radiation Oncology, University of Cincinnati and Cincinnati Children's Hospital, Cincinnati, Ohio
| | | | | | - Kathryn M Bondra
- Greehey Children's Cancer Research Institute, University of Texas, San Antonio, Texas
| | | | - Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas, San Antonio, Texas
| |
Collapse
|
17
|
Shirinbak S, Chan RY, Shahani S, Muthugounder S, Kennedy R, Hung LT, Fernandez GE, Hadjidaniel MD, Moghimi B, Sheard MA, Epstein AL, Fabbri M, Shimada H, Asgharzadeh S. Combined immune checkpoint blockade increases CD8+CD28+PD-1+ effector T cells and provides a therapeutic strategy for patients with neuroblastoma. Oncoimmunology 2021; 10:1838140. [PMID: 33489468 PMCID: PMC7801125 DOI: 10.1080/2162402x.2020.1838140] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Immune checkpoint therapy has resulted in minimal clinical response in many pediatric cancers. We sought to understand the influence of immune checkpoint inhibition using anti-PD-1 and anti-CTLA-4 antibodies individually, in combination, and after chemotherapy on immune responses in minimal and established murine neuroblastoma models. We also sought to understand the role of the tumor microenvironment (TME) and PD-L1 expression and their alteration post-chemotherapy in our models and human tissues. PD-L1 expression was enriched in human tumor-associated macrophages and up-regulated after chemotherapy. In a murine minimal disease model, single and dual immune checkpoint blockade promoted tumor rejection, improved survival, and established immune memory with long-term anti-tumor immunity against re-challenge. In an established tumor model, only dual immune checkpoint blockade showed efficacy. Interestingly, dual immune checkpoint therapy distinctly influenced adaptive and innate immune responses, with significant increase in CD8+CD28+PD-1+ T cells and inflammatory macrophages (CD11bhiCD11c−F4/80+Ly6Chi) in tumor-draining lymph nodes. Adding chemotherapy before immunotherapy provided significant survival benefit for mice with established tumors receiving anti-PD-1 or dual immune checkpoint blockade. Our findings demonstrate anti-PD-1 and anti-CTLA-4 therapy induces a novel subset of effector T cells, and support administration of induction chemotherapy immediately prior to immune checkpoint blockade in children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Soheila Shirinbak
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - Randall Y Chan
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA.,Department of Pediatrics, Los Angeles County + University of Southern California Medical Center, Los Angeles, CA, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shilpa Shahani
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sakunthala Muthugounder
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - Rebekah Kennedy
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - Long T Hung
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - G Esteban Fernandez
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - Michael D Hadjidaniel
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - Babak Moghimi
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael A Sheard
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA
| | - Alan L Epstein
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Muller Fabbri
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Hiroyuki Shimada
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shahab Asgharzadeh
- Department of Pediatrics, Children's Hospital Los Angeles and the Saban Research Institute, Los Angeles, CA, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Kushlinskii NE, Alferov AA, Timofeev YS, Gershtein ES, Bulycheva IV, Bondarev AV, Shchupak MY, Sokolov NY, Polikarpova SB, Efimova MM, Dzampaev AA, Sushentsov EA, Aliev MD, Musaev ER. Key Immune Checkpoint PD-1/PD-L1 Signaling Pathway Components in the Blood Serum from Patients with Bone Tumors. Bull Exp Biol Med 2020; 170:64-68. [PMID: 33231796 DOI: 10.1007/s10517-020-05005-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Indexed: 01/30/2023]
Abstract
The levels of sPD-1 and sPD-L1 were analyzed in blood serum of 132 patients (age 14-70 years) with primary bone tumors: osteosarcoma (N=39), chondrosarcoma (N=42), Ewing sarcoma (N=9), chordoma (N=12), giant-cell bone tumor (GCBT) (N=16), benign neoplasms (N=14) and in and practically healthy subjects (age 19-58 years; N=27). sPD-L1 levels in all studied bone neoplasms were significantly higher than in the control. Serum sPD-1 level in GCBT patients was significantly higher than in the control, benign neoplasms, chondrosarcoma, and chordoma patients, but did not differ from osteosarcoma group. sPD-1 concentration in Ewing sarcoma was significantly higher than in chordoma and chondrosarcoma, but did not differ from the control. sPD-1 level in chondrosarcoma patients was also lower than in osteosarcoma, Ewing sarcoma, and in the control. Both sPD-1 and sPD-L1 concentrations were not significantly associated with the type of affected bone, process localization, disease stage, tumor histological grade, patients' age and sex. These results suggest the possibility of using these biological markers for preliminary assessment of the character of the process in the bone.
Collapse
Affiliation(s)
- N E Kushlinskii
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A A Alferov
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yu S Timofeev
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E S Gershtein
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I V Bulycheva
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A V Bondarev
- Moscow City Oncological Hospital No. 62, Stepanovo, Moscow region, Russia
| | - M Yu Shchupak
- Moscow City Oncological Hospital No. 62, Stepanovo, Moscow region, Russia
| | - N Yu Sokolov
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - S B Polikarpova
- I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - M M Efimova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Dzampaev
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E A Sushentsov
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M D Aliev
- National Medical Research Center of Radiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E R Musaev
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
19
|
Abstract
Both the onset of various malignancies as well as the treatment of cancer can lead to neurologic symptoms which can be difficult to diagnose. In this review, we highlight the varied ways in which neurologic sequelae of cancer and its treatment manifest in children. Initial neurologic presentation may be secondary to mass effect or to immune-mediated paraneoplastic syndromes. Treatment effects on the nervous system may arise from surgery, chemotherapy, radiation, or bone marrow transplantation. In addition, the rapidly expanding field of immunotherapies for cancer has generated numerous new approaches to eradicating cancer including monoclonal antibodies, checkpoint inhibitors, and chimeric antigen receptor T cells (CAR-T cells), which have neurologic side effects mediated by immune responses that are also being recognized. Here we review common consult questions to the neurologist and our general approach to these scenarios including altered mental status, headaches, seizures, and sensorimotor complaints, considering the multifactorial nature of each.
Collapse
Affiliation(s)
- Caren Armstrong
- Department of Neurology, Johns Hopkins Hospital, 200 N Wolfe St Suite 2158, Baltimore, MD, 21287, USA
| | - Lisa R Sun
- Department of Neurology, Johns Hopkins Hospital, 200 N Wolfe St Suite 2158, Baltimore, MD, 21287, USA.
| |
Collapse
|
20
|
Thompson LL, Chang MS, McCormack L, Polyakov NJ, Song H, Huang JT, Chen ST. Discordance between recommended and delivered therapies for immunotherapy-associated cutaneous toxicities in pediatric populations. Pediatr Blood Cancer 2020; 67:e28653. [PMID: 32798306 DOI: 10.1002/pbc.28653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 11/08/2022]
Affiliation(s)
- Leah L Thompson
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael S Chang
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lindsay McCormack
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Boston Children's Hospital, Boston, Massachusetts
| | - Nicole J Polyakov
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hannah Song
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer T Huang
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Boston Children's Hospital, Boston, Massachusetts
| | - Steven T Chen
- Harvard Medical School, Boston, Massachusetts.,Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
21
|
Liu KX, Joshi S. "Re-educating" Tumor Associated Macrophages as a Novel Immunotherapy Strategy for Neuroblastoma. Front Immunol 2020; 11:1947. [PMID: 32983125 PMCID: PMC7493646 DOI: 10.3389/fimmu.2020.01947] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is the most common extracranial pediatric tumor and often presents with metastatic disease, and patients with high-risk neuroblastoma have survival rates of ~50%. Neuroblastoma tumorigenesis is associated with the infiltration of various types of immune cells, including myeloid derived suppressor cells, tumor associated macrophages (TAMs), and regulatory T cells, which foster tumor growth and harbor immunosuppressive functions. In particular, TAMs predict poor clinical outcomes in neuroblastoma, and among these immune cells, TAMs with an M2 phenotype comprise an immune cell population that promotes tumor metastasis, contributes to immunosuppression, and leads to failure of radiation or checkpoint inhibitor therapy. This review article summarizes the role of macrophages in tumor angiogenesis, metastasis, and immunosuppression in neuroblastoma and discusses the recent advances in "macrophage-targeting strategies" in neuroblastoma with a focus on three aspects: (1) inhibition of macrophage recruitment, (2) targeting macrophage survival, and (3) reprogramming of macrophages into an immunostimulatory phenotype.
Collapse
Affiliation(s)
- Kevin X. Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Shweta Joshi
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, UCSD Rady's Children's Hospital, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
22
|
Castellanos L, Tuffaha M, Koren D, Levitsky LL. Management of Diabetic Ketoacidosis in Children and Adolescents with Type 1 Diabetes Mellitus. Paediatr Drugs 2020; 22:357-367. [PMID: 32449138 DOI: 10.1007/s40272-020-00397-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetic ketoacidosis (DKA) is the end result of insulin deficiency in type 1 diabetes mellitus (T1D). Loss of insulin production leads to profound catabolism with increased gluconeogenesis, glycogenolysis, lipolysis, and muscle proteolysis causing hyperglycemia and osmotic diuresis. High levels of counter-regulatory hormones lead to enhanced ketogenesis and the release of 'ketone bodies' into the circulation, which dissociate to release hydrogen ions and cause an overwhelming acidosis. Dehydration, hyperglycemia, and ketoacidosis are the hallmarks of this condition. Treatment is effective repletion of insulin, fluids and electrolytes. Newer approaches to early diagnosis, treatment, and prevention may diminish the risk of DKA and its childhood complications including cerebral edema. However, the potential for some technical and pharmacologic advances in the management of T1D to increase DKA events must be recognized.
Collapse
Affiliation(s)
- Luz Castellanos
- Division of Pediatric Endocrinology and Pediatric Diabetes Center, Massachusetts General Hospital, 175 Cambridge Street, 5th Floor, Boston, MA, 02114, USA
| | - Marwa Tuffaha
- Division of Pediatric Endocrinology and Pediatric Diabetes Center, Massachusetts General Hospital, 175 Cambridge Street, 5th Floor, Boston, MA, 02114, USA
| | - Dorit Koren
- Division of Pediatric Endocrinology and Pediatric Diabetes Center, Massachusetts General Hospital, 175 Cambridge Street, 5th Floor, Boston, MA, 02114, USA
| | - Lynne L Levitsky
- Division of Pediatric Endocrinology and Pediatric Diabetes Center, Massachusetts General Hospital, 175 Cambridge Street, 5th Floor, Boston, MA, 02114, USA.
| |
Collapse
|
23
|
Guo F, Li S, Guo C, Xu X, Zhou X, Ma D, Cao Z, Bing Z, Cui Y. Circular RNA circMAGI3 accelerates the glycolysis of non-small cell lung cancer through miR-515-5p/HDGF. Am J Transl Res 2020; 12:3953-3963. [PMID: 32774748 PMCID: PMC7407684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/03/2020] [Indexed: 06/11/2023]
Abstract
The emerging roles of circular RNAs (circRNAs) in non-small cell lung cancer (NSCLC) have been convincingly proved. However, there are still numerous unknown circRNAs needing exploration. Here, present research performed a circRNA microarray analysis for the expression profile and identified a novel circRNA (circMAGI3, hsa_circ_0110498). Clinically, circMAGI3 was significantly up-regulated in NSCLC tissue and cells, which was closely correlated with unfavorable outcome for NSCLC patients. Functionally, circMAGI3 promoted the glycolysis and proliferation of NSCLC cells. Mechanistically, circMAGI3 functioned as a sponge for miR-515-5p to relieve its target gene HDGF expression, thereby accelerating the glycolysis of NSCLC. Collectively, this research identified the oncogenic role of circMAGI3 in the tumorigenesis through miR-515-5p/HDGF axis, providing a vital theoretical basis for treatment of NSCLC.
Collapse
Affiliation(s)
- Feng Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Xiaohui Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Xiaoyun Zhou
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Dongjie Ma
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Zhili Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| | - Yushang Cui
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100730, China
| |
Collapse
|
24
|
Terry RL, Meyran D, Ziegler DS, Haber M, Ekert PG, Trapani JA, Neeson PJ. Immune profiling of pediatric solid tumors. J Clin Invest 2020; 130:3391-3402. [PMID: 32538896 PMCID: PMC7324195 DOI: 10.1172/jci137181] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pediatric cancers, particularly high-risk solid tumors, urgently need effective and specific therapies. Their outlook has not appreciably improved in decades. Immunotherapies such as immune checkpoint inhibitors offer much promise, but most are only approved for use in adults. Though several hundred clinical trials have tested immune-based approaches in childhood cancers, few have been guided by biomarkers or clinical-grade assays developed to predict patient response and, ultimately, to help select those most likely to benefit. There is extensive evidence in adults to show that immune profiling has substantial predictive value, but few studies focus on childhood tumors, because of the relatively small disease population and restricted use of immune-based therapies. For instance, only one published study has retrospectively examined the immune profiles of pediatric brain tumors after immunotherapy. Furthermore, application and integration of advanced multiplex techniques has been extremely limited. Here, we review the current status of immune profiling of pediatric solid tumors, with emphasis on tumor types that represent enormous unmet clinical need, primarily in the context of immune checkpoint inhibitor therapy. Translating optimized and informative immune profiling into standard practice and access to personalized combination therapy will be critical if childhood cancers are to be treated effectively and affordably.
Collapse
Affiliation(s)
- Rachael L. Terry
- Children’s Cancer Institute, Randwick, New South Wales, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia
- Université de Paris, Inserm, U976 HIPI Unit, Institut de Recherche Saint-Louis, Paris, France
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - David S. Ziegler
- Children’s Cancer Institute, Randwick, New South Wales, Australia
- Kids Cancer Center, Sydney Children’s Hospital, Randwick, New South Wales, Australia
| | - Michelle Haber
- Children’s Cancer Institute, Randwick, New South Wales, Australia
| | - Paul G. Ekert
- Children’s Cancer Institute, Randwick, New South Wales, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia
| | - Joseph A. Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Bosse KR, Majzner RG, Mackall CL, Maris JM. Immune-Based Approaches for the Treatment of Pediatric Malignancies. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2020; 4:353-370. [PMID: 34113750 PMCID: PMC8189419 DOI: 10.1146/annurev-cancerbio-030419-033436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immune-based therapies have now been credentialed for pediatric cancers with the robust efficacy of chimeric antigen receptor (CAR) T cells for pediatric B cell acute lymphocytic leukemia (ALL), offering a chance of a cure for children with previously lethal disease and a potentially more targeted therapy to limit treatment-related morbidities. The developmental origins of most pediatric cancers make them ideal targets for immune-based therapies that capitalize on the differential expression of lineage-specific cell surface molecules such as antibodies, antibody-drug conjugates, or CAR T cells, while the efficacy of other therapies that depend on tumor immunogenicity such as immune checkpoint inhibitors has been limited to date. Here we review the current status of immune-based therapies for childhood cancers, discuss challenges to developing immunotherapeutics for these diseases, and outline future directions of pediatric immunotherapy discovery and development.
Collapse
Affiliation(s)
- Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Robbie G Majzner
- Department of Pediatrics and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Crystal L Mackall
- Department of Pediatrics and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
26
|
Ingley KM, Cohen-Gogo S, Gupta AA. Systemic therapy in pediatric-type soft-tissue sarcoma. ACTA ACUST UNITED AC 2020; 27:6-16. [PMID: 32174753 DOI: 10.3747/co.27.5481] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Soft-tissue sarcoma (sts) is rare and represents approximately 7% of cancers in children and in adolescents less than 20 years of age. Rhabdomyosarcoma (rms) is most prevalent in children less than 10 years of age and peaks again during adolescence (16-19 years of age). Multi-agent chemotherapy constitutes the mainstay of treatment for rms. In other non-rhabdomyosarcoma soft-tissue tumours, such as synovial sarcoma, evidence for routine use of chemotherapy is less robust, and alternative treatment options, including targeted agents and immunotherapy, are being explored. In this review, we focus on chemotherapy for pediatric-type rms and discuss the advances and challenges in systemic treatment for select non-rhabdomyosarcoma soft-tissue tumours in children and adolescents. We support an increasingly cooperative approach for treating pediatric and adult sts.
Collapse
Affiliation(s)
- K M Ingley
- Department of Pediatric Oncology, Royal Children's Hospital, Melbourne, Australia.,Adolescent and Young Adult Cancer Service, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - S Cohen-Gogo
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON
| | - A A Gupta
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON.,Division of Medical Oncology and Hematology, Sinai Health System, Toronto, ON
| |
Collapse
|
27
|
Shemesh CS, Chanu P, Jamsen K, Wada R, Rossato G, Donaldson F, Garg A, Winter H, Ruppel J, Wang X, Bruno R, Jin J, Girish S. Population pharmacokinetics, exposure-safety, and immunogenicity of atezolizumab in pediatric and young adult patients with cancer. J Immunother Cancer 2019; 7:314. [PMID: 31753029 PMCID: PMC6868826 DOI: 10.1186/s40425-019-0791-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The iMATRIX-atezolizumab study was a phase I/II, multicenter, open-label study designed to assess the safety and pharmacokinetics of atezolizumab in pediatric and young adult patients. We describe the pharmacokinetics (PK), exposure-safety, and immunogenicity of atezolizumab in pediatric and young adults with metastatic solid tumors or hematologic malignancies enrolled in this study. METHODS Patients aged < 18 years (n = 69) received a weight-adjusted dose of atezolizumab (15 mg/kg every 3 weeks [q3w]; maximum 1200 mg); those aged ≥ 18 years (n = 18) received a flat dose (1200 mg q3w). A prior two-compartment intravenous infusion input adult population-PK (popPK) model of atezolizumab was used as a basis to model pediatric data. RESULTS A total of 431 atezolizumab serum concentrations from 87 relapse-refractory pediatric and young adult patients enrolled in the iMATRIX-atezolizumab study were used for the popPK analysis. The dataset comprised predominantly patients aged < 18 years, including two infants aged < 2 years, with a wide body weight and age range. The clearance and volume of distribution estimates of atezolizumab were 0.217 L/day and 3.01 L, respectively. Atezolizumab geometric mean trough exposures were ~ 20% lower in pediatric patients versus young adults; this was not clinically meaningful as both groups achieved the target concentration (6 μg/mL). Safety was similar between pediatric and young adult patients with no exposure-safety relationship observed. Limited responses (4/87) precluded an exposure-response assessment on outcomes. A comparable rate (13% vs 11%) of atezolizumab anti-drug antibodies was seen in pediatric and young adult patients. CONCLUSIONS These findings demonstrate a similar exposure-safety profile of atezolizumab in pediatric and young adult patients, supportive of weight-based dosing in pediatric patients. TRIAL REGISTRATION NCT02541604.
Collapse
MESH Headings
- Adolescent
- Adult
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/blood
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/therapeutic use
- Child
- Child, Preschool
- Female
- Humans
- Infant
- Male
- Models, Biological
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/mortality
- Progression-Free Survival
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- Colby S Shemesh
- Department of Clinical Pharmacology Oncology, Genentech Inc., South San Francisco, CA, 94080, USA.
| | - Pascal Chanu
- Clinical Pharmacology, Modeling and Simulation, Genentech/Roche, Marseille, France
| | - Kris Jamsen
- Certara Strategic Consulting, Princeton, NJ, USA
| | - Russ Wada
- Certara Strategic Consulting, Princeton, NJ, USA
| | | | | | - Amit Garg
- Department of Clinical Pharmacology Oncology, Genentech Inc., South San Francisco, CA, 94080, USA
- Present address: Quantitative Pharmacology and Disposition, Seattle Genetics, South San Francisco, CA, USA
| | - Helen Winter
- Department of Clinical Pharmacology Oncology, Genentech Inc., South San Francisco, CA, 94080, USA
- Present address: Quantitative Pharmacology and Disposition, Seattle Genetics, South San Francisco, CA, USA
| | - Jane Ruppel
- Bioanalytical Sciences, Genentech Inc., South San Francisco, CA, USA
| | - Xin Wang
- Department of Clinical Pharmacology Oncology, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Rene Bruno
- Clinical Pharmacology, Modeling and Simulation, Genentech/Roche, Marseille, France
| | - Jin Jin
- Department of Clinical Pharmacology Oncology, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Sandhya Girish
- Department of Clinical Pharmacology Oncology, Genentech Inc., South San Francisco, CA, 94080, USA
| |
Collapse
|
28
|
Ihara K. Immune checkpoint inhibitor therapy for pediatric cancers: A mini review of endocrine adverse events. Clin Pediatr Endocrinol 2019; 28:59-68. [PMID: 31384097 PMCID: PMC6646237 DOI: 10.1297/cpe.28.59] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/15/2019] [Indexed: 12/27/2022] Open
Abstract
In recent years, immune checkpoint inhibitor therapy has attracted a great deal of attention in the field of cancer treatment. In the clinical setting, antibodies targeting programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have been successfully used to treat adult patients with various types of intractable cancer. However, in a substantial number of patients, ICI therapy is associated with autoimmune toxicities known as immune-related adverse events (IRAEs). Endocrinopathies, such as hypophysitis or autoimmune thyroid disease, may occur and can present unique clinical features that have not been documented with traditional chemotherapies. A Japanese clinical trial evaluating the anti-PD-1 antibody nivolumab for the treatment of pediatric patients with refractory malignant solid tumors and Hodgkin lymphoma has been ongoing since 2017. Moreover, tumors associated with Lynch syndrome, a hereditary form of mismatch repair deficiency, are being focused and represent the next target for ICI therapy in Japan. For the safe management of pediatric cancer patients treated with ICIs, pediatric endocrinologists must be aware of the risk of autoimmune endocrinopathies and perform relevant screening tests at appropriate stages of growth and development.
Collapse
Affiliation(s)
- Kenji Ihara
- Department of Pediatrics, Oita University Faculty of Medicine, Yufu, Japan
| |
Collapse
|