1
|
Krishna S, Prajapati B, Seth P, Sinha S. Dickopff 1 inhibits cancer stem cell properties and promotes neuronal differentiation of human neuroblastoma cell line SH-SY5Y. IBRO Neurosci Rep 2024; 17:73-82. [PMID: 39021664 PMCID: PMC11253693 DOI: 10.1016/j.ibneur.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/24/2024] [Indexed: 07/20/2024] Open
Abstract
Neuroblastomas are pediatric tumors arising from undifferentiated cells of neural crest origin with stem cell-like characteristics. Dysregulation of Wnt/β-catenin signaling has been shown to be linked to the development of various tumors. Activated Wnt signaling results in β-catenin accumulation in the nucleus to support pro-neoplastic traits. DKK1, a secreted glycoprotein, is an inhibitor of Wnt signaling, and the addition of DKKI to the culture medium has been used to suppress the Wnt pathway. This study aimed to analyze the role of Dickopff-1 as a potential differentiating agent for the neuroblastoma cell line SH-SY5Y and neurospheres derived from it. The treatment of SH-5Y5Y derived neurospheres by DKK1 resulted in their disintegration and reduced proliferation markers like Ki67, PCNA. DKK1 treatment to the neurospheres also resulted in the loss of cancer stem cell markers like CD133, KIT and pluripotency markers like SOX2, OCT4, NANOG. DKK1 treatment caused reduction in mRNA expression of β-catenin and TCF genes like TCF4, TCF12. When the SH-SY5Y cancer cells were grown under differentiating conditions, DKKI caused neuronal differentiation by itself, and in synergy with retinoic acid. This was verified by the expression of markers like MAPT, DCX, GAP43, ENO2 and also with changes in neurite length. We concluded that Wnt inhibition, as exemplified by DKK1 treatment, is therefore a possible differentiating condition and also suppresses the proliferative and cancer stemness related properties of SH-SY5Y neuroblastoma cells.
Collapse
Affiliation(s)
| | - Bharat Prajapati
- National Brain Research Centre, Manesar, Gurugram, India
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, Gothenburg, Sweden
| | - Pankaj Seth
- National Brain Research Centre, Manesar, Gurugram, India
| | - Subrata Sinha
- National Brain Research Centre, Manesar, Gurugram, India
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
2
|
Chen L, Chen D, Pan Y, Mo Y, Lai B, Chen H, Zhang DW, Xia XD. Inhibition of mitochondrial OMA1 ameliorates osteosarcoma tumorigenesis. Cell Death Dis 2024; 15:786. [PMID: 39487118 PMCID: PMC11530700 DOI: 10.1038/s41419-024-07127-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 11/04/2024]
Abstract
OMA1 is an ATP-independent zinc metalloprotease essential for maintaining mitochondrial homeostasis and plays a vital role in tumorigenesis. Depending on the type of cancer, a decrease in OMA1 expression has been linked to a varying prognosis for patients. The role of OMA1 in human osteosarcoma (OS), one of the most prevalent malignant bone tumors, remains elusive. Here, we observed elevated OMA1 expression in OS tumor tissues from four patients with advanced OS. Knockout of OMA1 in OS cells significantly reduces OS tumor weight and size, and lung metastatic nodules in BALB/c nude mice. Immunohistochemistry analysis showed a significant decrease in Ki67 and an increase in Cleaved-caspase 3 in OMA1 knockout tumor samples. Mechanistically, we found that OMA1 deficiency increases the levels of PINK1 and Parkin and consequently induces excessive mitophagy, leading to increased apoptosis and reduced cell proliferation and invasion in OS cells. Specifically, OMA1 deficiency reduces the amount of cytosolic p53 and p53-associated cytosolic Parkin but increases mitochondrial p53, which may lead to enhanced apoptosis. Regarding the effect on cell proliferation and invasion, loss of OMA1 reduces mitochondrial ROS levels and increases cytosolic glycogen synthase kinase 3β (GSK3β) levels, thereby increasing interaction between GSK3β and β-catenin and then reducing cytosolic and nuclear β-catenin. This contributes to reduced cell proliferation and migration in OMA1-deficient cells. Moreover, we found that ciclopirox (CPX), an antifungal drug, induces OMA1 self-cleavage and L-OMA1 degradation in cultured OS cells. CPX also reduces tumor development of control OS cells but not OMA1-deficient OS cells in mice. These findings strongly support the important role of OMA1 in OS tumorigenesis and suggest that OMA1 may be a valuable prognostic marker and a promising therapeutic target for OS.
Collapse
Affiliation(s)
- Lingyan Chen
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China.
| | - Dejian Chen
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Yiming Pan
- Department of Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yimei Mo
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Biyu Lai
- Department of Radiology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Huiguang Chen
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Da-Wei Zhang
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| | - Xiao-Dan Xia
- Department of Orthopedics, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China.
| |
Collapse
|
3
|
Zhou O, Harila A, Hovén E, Lönnerblad M. Educational outcomes school year nine in children treated for acute lymphoblastic leukemia: A nationwide registry-based study from Sweden. Int J Cancer 2024. [PMID: 39425561 DOI: 10.1002/ijc.35231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Acute lymphoblastic leukemia (ALL) constitutes approximately 25% of pediatric cancers, and with contemporary protocols, the 5-year survival rate is over 90%. Despite improved survival, neurocognitive impairments from treatment raise concerns. This registry study aimed to explore the impact of ALL treatment on educational outcomes from school year nine in Swedish children. A population-based cohort of 503 children diagnosed with ALL from 1990 to 2010 was identified from the Swedish Childhood Cancer Registry and matched with five controls each. Assessed variables were delayed graduation, high school eligibility, total merit value, school grades in Swedish, English, mathematics, and physical education, and results in national tests. Analyses were performed between cases and controls and by sex, age at diagnosis, and risk group. Our results showed that, compared to controls, cases had higher odds for delayed graduation, poorer results in physical education, and higher rates of absence in national tests in English and mathematics. Children in the standard-risk group (treated with first-line chemotherapy only) exhibited similar results to matched controls whereas children in the high-risk group (treated with cranial irradiation, hematological stem cell transplantation, or/and for ALL relapse and thus likely received also radiotherapy) had lower total merit value compared to controls. We conclude that Swedish children diagnosed with ALL between the years 1990-2010 mainly exhibited comparable educational outcomes to controls, although children in the high-risk group had lower results. These findings highlight the importance of evaluating especially children with high-risk ALL in order to identify those requiring educational support and for designing targeted interventions.
Collapse
Affiliation(s)
- Otto Zhou
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Arja Harila
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Emma Hovén
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Malin Lönnerblad
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Special Education, Stockholm University, Stockholm, Sweden
| |
Collapse
|
4
|
Wells ME, Eckhoff MD, Davis W, Singh V, Rajani R, Polfer EM. Ewing Sarcoma in the Pediatric Population: Predictors of Survival Within the United States. J Am Acad Orthop Surg Glob Res Rev 2024; 8:01979360-202410000-00017. [PMID: 39436736 PMCID: PMC11498927 DOI: 10.5435/jaaosglobal-d-24-00281] [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: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Bone and joint tumors are the third most common cause of pediatric cancer-related deaths in the United States. Although there have been improvements in survival rates among pediatric cancer patients over the past few decades, bone and joint cancers remain the exception. Considering current clinical trials involving novel targeted therapies, the establishment of updated mortality rates and predictors of survival for this cancer would be prudent. This investigation sought to determine updated 5-year survival rates and predictors of survival among pediatric Ewing sarcoma (ES) of bone treated within the United States. METHODS The National Cancer Database was retrospectively inquired for all pediatric ES cases within the most updated bone and joint public use file available in September 2022. The reported data were truncated to only include patients with reported 5-year vital (ie, survival) status. Cox proportional hazard regression was conducted on both the truncated data and the entire cohort to validate the findings. The patients were then separated into alive versus deceased cohorts, and univariate regression analysis was done followed by multivariable regression of notable variables of interest. RESULTS Overall, an aggregated 5-year survival rate of 74.5% was found in the included patient cohort. Patients with localized cancer had a comparatively improved 5-year survival rate of 84.70% as opposed to those with macrometastatic disease on presentation with a survival rate of 50.4%. Patient demographic-, tumor-, and treatment-specific variables all demonstrated an effect on survival. The multivariable predictors of worse mortality were found to include older age, larger tumor size (>8 cm), macrometastatic disease on presentation, and positive surgical margins. CONCLUSION This analysis serves to establish updated survival rates of pediatric ES treated within the United States to set standards for comparison among future studies. Continued multi-institutional and international collaboration is needed to optimize current treatment results and develop novel targeted therapies.
Collapse
Affiliation(s)
- Matthew E. Wells
- From the Department of Orthopedic Surgery, William Beaumont Army Medical Center (Dr. Wells, Dr. Eckhoff, and Dr. Polfer); the Department of Orthopedic Surgery, Texas Tech University Health Sciences Center El Paso (Dr. Wells, Dr. Eckhoff, Dr. Davis, Dr. Rajani, and Dr. Polfer); and Biostatistics and Epidemiology Consulting Lab, Texas Tech University Health Sciences Center El Paso, El Paso, TX (Dr. Singh)
| | - Michael D. Eckhoff
- From the Department of Orthopedic Surgery, William Beaumont Army Medical Center (Dr. Wells, Dr. Eckhoff, and Dr. Polfer); the Department of Orthopedic Surgery, Texas Tech University Health Sciences Center El Paso (Dr. Wells, Dr. Eckhoff, Dr. Davis, Dr. Rajani, and Dr. Polfer); and Biostatistics and Epidemiology Consulting Lab, Texas Tech University Health Sciences Center El Paso, El Paso, TX (Dr. Singh)
| | - William Davis
- From the Department of Orthopedic Surgery, William Beaumont Army Medical Center (Dr. Wells, Dr. Eckhoff, and Dr. Polfer); the Department of Orthopedic Surgery, Texas Tech University Health Sciences Center El Paso (Dr. Wells, Dr. Eckhoff, Dr. Davis, Dr. Rajani, and Dr. Polfer); and Biostatistics and Epidemiology Consulting Lab, Texas Tech University Health Sciences Center El Paso, El Paso, TX (Dr. Singh)
| | - Vishwajeet Singh
- From the Department of Orthopedic Surgery, William Beaumont Army Medical Center (Dr. Wells, Dr. Eckhoff, and Dr. Polfer); the Department of Orthopedic Surgery, Texas Tech University Health Sciences Center El Paso (Dr. Wells, Dr. Eckhoff, Dr. Davis, Dr. Rajani, and Dr. Polfer); and Biostatistics and Epidemiology Consulting Lab, Texas Tech University Health Sciences Center El Paso, El Paso, TX (Dr. Singh)
| | - Rajiv Rajani
- From the Department of Orthopedic Surgery, William Beaumont Army Medical Center (Dr. Wells, Dr. Eckhoff, and Dr. Polfer); the Department of Orthopedic Surgery, Texas Tech University Health Sciences Center El Paso (Dr. Wells, Dr. Eckhoff, Dr. Davis, Dr. Rajani, and Dr. Polfer); and Biostatistics and Epidemiology Consulting Lab, Texas Tech University Health Sciences Center El Paso, El Paso, TX (Dr. Singh)
| | - Elizabeth M. Polfer
- From the Department of Orthopedic Surgery, William Beaumont Army Medical Center (Dr. Wells, Dr. Eckhoff, and Dr. Polfer); the Department of Orthopedic Surgery, Texas Tech University Health Sciences Center El Paso (Dr. Wells, Dr. Eckhoff, Dr. Davis, Dr. Rajani, and Dr. Polfer); and Biostatistics and Epidemiology Consulting Lab, Texas Tech University Health Sciences Center El Paso, El Paso, TX (Dr. Singh)
| |
Collapse
|
5
|
Chen H, Wang W, Chang S, Huang X, Wang N. A useful mTORC1 signaling-related RiskScore model for the personalized treatment of osteosarcoma patients by using the bulk RNA-seq analysis. Discov Oncol 2024; 15:418. [PMID: 39251459 PMCID: PMC11383908 DOI: 10.1007/s12672-024-01301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
AIMS This research developed a prognostic model for OS patients based on the Mechanistic Target of Rapamycin Complex 1 (mTORC1) signature. BACKGROUND The mTORC1 signaling pathway has a critical role in the maintenance of cellular homeostasis and tumorigenesis and development through the regulation of cell growth, metabolism and autophagy. However, the mechanism of action of this signaling pathway in Osteosarcoma (OS) remains unclear. OBJECTIVE The datasets including the TARGET-OS and GSE39058, and 200 mTORC1 genes were collected. METHODS The mTORC1 signaling-related genes were obtained based on the Molecular Signatures Database (MSigDB) database, and the single sample gene set enrichment analysis (ssGSEA) algorithm was utilized in order to calculate the mTORC1 score. Then, the WGCNA were performed for the mTORC1-correlated gene module, the un/multivariate and lasso Cox regression analysis were conducted for the RiskScore model. The immune infiltration analysis was performed by using the ssGSEA method, ESTIMATE tool and MCP-Count algorithm. KM survival and Receiver Operating Characteristic (ROC) Curve analysis were performed by using the survival and timeROC package. RESULTS The mTORC1 score and WGCNA with β = 5 screened the mTORC1 positively correlated skyblue2 module that included 67 genes, which are also associated with the metabolism and hypoxia pathways. Further narrowing of candidate genes and calculating the regression coefficient, we developed a useful and reliable RiskScore model, which can classify the patients in the training and validation set into high and low-risk groups based on the median value of RiskScore as an independent and robust prognostic factor. High-risk patients had a significantly poor prognosis, lower immune infiltration level of multiple immune cells and prone to cancer metastasis. Finally, we a nomogram model incorporating the metastasis features and RiskScore showed excellent prediction accuracy and clinical practicability. CONCLUSION We developed a useful and reliable risk prognosis model based on the mTORC1 signaling signature.
Collapse
Affiliation(s)
- Hongxia Chen
- Department of Hematology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Wei Wang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Shichuan Chang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Xiaoping Huang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China.
| | - Ning Wang
- Department of Oncology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China.
| |
Collapse
|
6
|
Pujol Manresa A, Buendía López S, Andión M, Herrero B, Lassaletta Á, Ramirez M, Ruano D, Hernández-Marqués C, Varo A, de Rojas T, Cortés Hernández M, Verdú-Amorós J, Martín Prado S, Artigas A, Redondo E, Ruiz Pato J, Herreros López P, Sevilla J, Madero L, Moreno L, Bautista Sirvent F, Rubio-San-Simón A. Safety and outcome of children, adolescents and young adults participating in phase I/II clinical oncology trials: a 9-year center experience. Front Pediatr 2024; 12:1423484. [PMID: 39318620 PMCID: PMC11421171 DOI: 10.3389/fped.2024.1423484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/13/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Enrolling children with cancer in early phase trials is crucial to access innovative treatments, contributing to advancing pediatric oncology research and providing tailored therapeutic options. Our objective is to analyze the impact of these trials on patient outcomes and safety, and to examine the evolution and feasibility of trials in pediatric cancer over the past decade. Methods All patients recruited in pediatric anticancer phase I/II clinical trials from January 2014 to December 2022 were included. Clinical records and trial protocols were analyzed. Results A total of 215 patients (median age 11.2 years, range 1-29.5) were included in 52 trials (258 inclusions). Patients with extracranial solid tumors (67%), central nervous system (CNS) tumors (24%), and leukemia (9%) were included. The most common investigational drugs were small molecules (28.3%) and antibodies (20.5%). Serious adverse events were experienced by 41% of patients, 4.4% discontinued treatment because of toxicity and two had toxic deaths. Median event-free survival was 3.7 months (95%CI: 2.8-4.5), longer in phase II trials than in phase I (2 vs. 6.3 months; p ≤ 0.001). Median overall survival was 12 months (95%CI: 9-15), higher in target-specific vs. non-target-specific trials (14 vs. 6 months; p ≤ 0.001). Discussion A significant and increasing number of patients have been included in early clinical trials, suggesting that both oncologists and families consider it valuable to be referred to specialized Units to access new therapies. Moreover, our data suggests that participation in early clinical trials, although not without potential toxicities, might have a positive impact on individual outcomes.
Collapse
Affiliation(s)
- Anna Pujol Manresa
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Susana Buendía López
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Maitane Andión
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Health, Madrid, Spain
| | - Blanca Herrero
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Health, Madrid, Spain
| | - Álvaro Lassaletta
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Manuel Ramirez
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Health, Madrid, Spain
| | - David Ruano
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Health, Madrid, Spain
| | - Carmen Hernández-Marqués
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Amalia Varo
- Pediatric Hematology-Oncology Department, Pediatric Cancer Center Barcelona, Barcelona, Spain
| | | | - Marta Cortés Hernández
- Pediatric Hematology-Oncology Department, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Jaime Verdú-Amorós
- Pediatric Hematology-Oncology Department, Hospital Clínico Universitario, Valencia, Spain
- Biomedical Research Institute, INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Silvia Martín Prado
- Pharmacy Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Andrea Artigas
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Esther Redondo
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Julia Ruiz Pato
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Pilar Herreros López
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Julián Sevilla
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Luis Madero
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Health, Madrid, Spain
| | - Lucas Moreno
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Francisco Bautista Sirvent
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Trial and Data Centrum, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Alba Rubio-San-Simón
- Pediatric Hematology-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Health, Madrid, Spain
| |
Collapse
|
7
|
Boensvang NN, Weibel M, Wakefield CE, Bidstrup PE, Olsen M, Nissen KB, Spager V, Fridh MK, Larsen HB. Online Ambassador Visits for Hospitalized Children With Cancer: Qualitative Evaluation of Implementation. JMIR Pediatr Parent 2024; 7:e53309. [PMID: 39231427 PMCID: PMC11411222 DOI: 10.2196/53309] [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: 10/03/2023] [Revised: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Children with cancer or cancer-like disease risk treatment-related isolation, which can negatively impact their peer relationships and social competencies and exacerbate their loneliness. During the COVID-19 pandemic, increased online socialization became the new normal imposed by national isolation guidelines. To adhere to the treatment-related isolation guidelines, children with cancer were offered online classmate "ambassador" visits during hospitalization. OBJECTIVE This study aimed to identify facilitators and barriers to online classmate "ambassador" visits during children with cancer's hospitalization through a qualitative descriptive process evaluation using the Consolidated Framework for Implementation Research. METHODS From January to April 2022, we conducted 39 individual semistructured interviews with hospitalized children (n=16), their classmates (n=16), teachers from their schools (n=3), and study nurses (n=4) from involved hospitals. Most interviews (n=37, 95%) were conducted online using Microsoft Teams or Google Meet, while 2 (5%) interviews were conducted in person at the participants' residences. This approach allowed us to gain a broad understanding of the facilitators and barriers to online ambassador visits. RESULTS We identified four themes: (1) working together, (2) ensuring participation, (3) staying connected, and (4) together online. The themes are described in terms of facilitators and barriers to online ambassador visits with 3 Consolidated Framework for Implementation Research domains: innovation, individuals, and the implementation process. CONCLUSIONS Addressing the social needs of hospitalized children through online visits with their classmates may be relevant when one-on-one meetings are problematic. The online visits are highly dependent on collaboration between study nurses and teachers and assessing the needs of the hospitalized children. While a high degree of adult engagement and a stable internet connection are pivotal, these online visits can promote much-needed social interaction between children across physical settings.
Collapse
Affiliation(s)
- Natasha Nybro Boensvang
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Paediatrics and Adolescent Medicine, University Hospital of Copehagen (Rigshospitalet), Copenhagen, Denmark
| | - Mette Weibel
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Paediatrics and Adolescent Medicine, University Hospital of Copehagen (Rigshospitalet), Copenhagen, Denmark
| | - Claire E Wakefield
- Behavioral Sciences Unit, Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
- School for Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Pernille Envold Bidstrup
- Psychological Aspects of Cancer, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Olsen
- Department of Paediatrics and Adolescent Medicine, Paediatric Haematology and Oncology Section, University Hospital Aalborg, Aalborg, Denmark
| | - Karin Bækgaard Nissen
- Department of Paediatric and Adolescent Medicine, University Hospital Aarhus, Aarhus, Denmark
| | - Vibeke Spager
- Department of Paediatrics and Adolescent Medicine, University Hospital of Copehagen (Rigshospitalet), Copenhagen, Denmark
| | - Martin Kaj Fridh
- Department of Paediatrics and Adolescent Medicine, University Hospital of Copehagen (Rigshospitalet), Copenhagen, Denmark
| | - Hanne Bækgaard Larsen
- Department of Paediatrics and Adolescent Medicine, University Hospital of Copehagen (Rigshospitalet), Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
- The Paediatric Clinic, Juliane Marie Centre, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Du XX, Todorov P, Isachenko E, Sanchez R, Uribe P, Rahimi G, Mallmann P, Isachenko V. Model of micro-metastases of breast cancer cells in ovarian tissue: Cryopreservation of ZR-75-1 and MDA-MB-231 cells with increased speed of warming increases malignancy. Cryobiology 2024; 116:104910. [PMID: 38777075 DOI: 10.1016/j.cryobiol.2024.104910] [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: 07/24/2023] [Revised: 04/16/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
In medicine, ovarian tissue cryopreservation exists for fertility preservation of cancer patients. In fact, ovarian tissue frozen for subsequent thawing and re-transplantation can be contaminated with cancer cells. Therefore, investigations on the effect of cryopreservation on the post-thawed viability of such cells are relevant. Speed of warming is a key parameter of cell cryopreservation. However, the data about comparative viability of cancer cells cryopreserved with different parameters of warming are limited. The aim of our investigations was to assess the malignancy of cryopreserved cancer cells after conventional cooling followed by relatively slow and quick speed of warming. In vitro cultured breast cancer cells of lines ZR-75-1 and MD0MD-231 in form of compacted fragments (as a model of solid tumors) were frozen following a protocol usually used for freezing of ovarian tissue (6 % ethylene glycol+6 % glycerol+0.15 M sucrose, -0.3 °C/min). Cells were warmed by two routine regimes of warming: at 37 °C ("slow" warming) and 100 °C ("quick" warming). Biological properties of cells were investigated: viability, proliferation rate, 2D- and 3D-migration, transmembrane movement and invasion. Quick warming at 100 °C in comparison with slow warming at 37 °C exhibited significantly higher cell survival for MDA-MB-231 cells: 70.1 % vs. 63.2 % and for ZR-75-1 86.8 % vs. 82.9 %, respectively. The cell motility including 2D movement and 3D transmembrane migration were higher after quick thawing at 100 °C. Invasive abilities of cells after cryopreservation were higher than that of fresh (non-treated cells). Both thawing regimes showed a similar rate of cell proliferation. Cryopreservation procedures, and especially this one with quick thawing, increase malignancy of ZR-75-1 and MDA-MB-231 breast cancer cells and risk of metastasis.
Collapse
Affiliation(s)
- Xin-Xin Du
- Research Group for Reproductive Medicine, IVF-Laboratory and Department of Gynecology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Plamen Todorov
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences (BAS), Sofia, Bulgaria.
| | - Evgenia Isachenko
- Research Group for Reproductive Medicine, IVF-Laboratory and Department of Gynecology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Raul Sanchez
- Temuco University de la Frontera, Temuco, Chile.
| | - Pamela Uribe
- Temuco University de la Frontera, Temuco, Chile.
| | - Gohar Rahimi
- Research Group for Reproductive Medicine, IVF-Laboratory and Department of Gynecology, Medical Faculty, University of Cologne, Cologne, Germany; Medizinisches Versorgungszentrum AMEDES für IVF- und Pränatalmedizin in Köln GmbH, Cologne, Germany.
| | - Peter Mallmann
- Research Group for Reproductive Medicine, IVF-Laboratory and Department of Gynecology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Volodimir Isachenko
- Research Group for Reproductive Medicine, IVF-Laboratory and Department of Gynecology, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
9
|
Chang YH, Yu CH, Lu MY, Jou ST, Lin CY, Lin KH, Chang HH, Ni YL, Chou SW, Ko KY, Lin DT, Hsu WM, Chen HY, Yang YL. Higher tumor mutational burden is associated with inferior outcomes among pediatric patients with neuroblastoma. Pediatr Blood Cancer 2024; 71:e31176. [PMID: 38967585 DOI: 10.1002/pbc.31176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/18/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Neuroblastoma is a pediatric malignancy with heterogeneous clinical outcomes. Our aim was to identify prognostic genetic markers for patients with neuroblastoma, who were treated with the Taiwan Pediatric Oncology Group (TPOG) neuroblastoma N2002 protocol, to improve risk stratification and inform treatment. METHODS Our analysis was based on 53 primary neuroblastoma specimens, diagnosed pre-chemotherapy, and 11 paired tumor relapse specimens. Deep sequencing of 113 target genes was performed using a custom panel. Multiplex ligation-dependent probe amplification was performed to identify clinical outcomes related to copy-number variations. RESULTS We identified 128 variations associated with survival, with the number of variations being higher in the relapse than that in the diagnostic specimen (p = .03). The risk of event and mortality was higher among patients with a tumor mutational burden ≥10 than that in patients with a lower burden (p < .0001). Multivariate analysis identified tumor mutational burden, MYCN amplification, and chromosome 3p deletion as significant prognostic factors, independent of age at diagnosis, sex, and tumor stage. The 5-year event-free survival and overall survival rate was lower among patients with high tumor burden than in patients with low tumor burden. Furthermore, there was no survival of patients with an ALK F1147L variation at 5 years after diagnosis. CONCLUSIONS Genome sequencing to determine the tumor mutational burden and ALK variations can improve the risk classification of neuroblastoma and inform treatment.
Collapse
Affiliation(s)
- Ya-Hsuan Chang
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Chih-Hsiang Yu
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
- Departments of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Meng-Yao Lu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shiann-Tarng Jou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Yu Lin
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Kai-Hsin Lin
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsiu-Hao Chang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ling Ni
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Wei Chou
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuan-Yin Ko
- Department of Nuclear Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dong-Tsamn Lin
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Yu Chen
- Institute of Statistical Science Academia Sinica, Taipei, Taiwan
| | - Yung-Li Yang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine and Medical Service, National Taiwan University Cancer Center, Taipei, Taiwan
| |
Collapse
|
10
|
Westerveld ASR, Roesthuis P, van der Pal HJH, Bresters D, Bierings M, Loonen J, de Vries ACH, Louwerens M, Koopman MMW, van den Heuvel-Eibrink MM, van der Heiden-van der Loo M, Hoogerbrugge P, Janssens GO, de Krijger RR, Ronckers CM, Pieters R, Kremer LCM, Teepen JC. Increased risk of subsequent neoplasm after hematopoietic stem cell transplantation in 5-year survivors of childhood acute lymphoblastic leukemia. Blood Cancer J 2024; 14:150. [PMID: 39198413 PMCID: PMC11358316 DOI: 10.1038/s41408-024-01122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) survivors are at risk for developing subsequent neoplasms, but there is limited information on long-term risks and risk factors for both subsequent malignant neoplasms (SMNs) and subsequent non-malignant neoplasms (SNMNs). We analyzed long-term risk and risk factors for SMNs and SNMNs among 3291 5-year ALL survivors from the Dutch Childhood Cancer Survivor Study-LATER cohort (1963-2014). We calculated standardized incidence ratios (SIRs) and cumulative incidences and used multivariable Cox proportional hazard regression analyses for analyzing risk factors. A total of 97 survivors developed SMNs and 266 SNMNs. The 30-year cumulative incidence was 4.1% (95%CI: 3.5-5.3) for SMNs and 10.4%(95%CI: 8.9-12.1) for SNMNs. Risk of SMNs was elevated compared to the general population (SIR: 2.6, 95%CI: 2.1-3.1). Survivors treated with hematopoietic stem cell transplantation (HSCT) with total body irradiation (TBI) (HR:4.2, 95%CI: 2.3-7.9), and without TBI (HR:4.0,95%CI: 1.2-13.7) showed increased SMN risk versus non-transplanted survivors. Cranial radiotherapy (CRT) was also a risk factor for SMNs (HR:2.1, 95%CI: 1.4-4.0). In conclusion, childhood ALL survivors have an increased SMN risk, especially after HSCT and CRT. A key finding is that even HSCT-treated survivors without TBI treatment showed an increased SMN risk, possibly due to accompanied chemotherapy treatment. This emphasizes the need for careful follow-up of HSCT and/or CRT-treated survivors.
Collapse
Affiliation(s)
| | - Pien Roesthuis
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Dorine Bresters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marc Bierings
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Andrica C H de Vries
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Erasmus Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Maria M W Koopman
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | | | | | - Geert O Janssens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cecile M Ronckers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division of Childhood Cancer Epidemiology (EpiKiK), Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Leontien C M Kremer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | - Jop C Teepen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
11
|
Wei X, Bjarkadottir BD, Nadjaja D, Sheikh S, Fatum M, Lane S, Williams SA. Effect of AMH on primordial follicle populations in mouse ovaries and human pre-pubertal ovarian xenografts during doxorubicin treatment. Front Cell Dev Biol 2024; 12:1449156. [PMID: 39258229 PMCID: PMC11383774 DOI: 10.3389/fcell.2024.1449156] [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: 06/14/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Survival rates of the childhood cancer patients are improving, however cancer treatments such as chemotherapy may lead to infertility due to loss of the primordial follicle (PMF) reserve. Doxorubicin (DXR) is a gonadotoxic chemotherapy agent commonly used in childhood cancers. Anti-Müllerian Hormone (AMH) has been reported to have a protective effect on the mouse ovarian reserve against DXR in vivo. However, whether AMH can prevent PMF loss in conjunction with DXR in human ovarian tissue in vivo has not been determined. Methods In order to investigate this, we first established an optimum dose of DXR that induced PMF loss in cultured mouse ovaries and investigated the efficacy of AMH on reducing DXR-induced PMF loss in mice in vitro. Second, we investigated the effects of DXR on pre-pubertal human ovarian tissue and the ability of AMH to prevent DXR-induced damage comparing using a mouse xenograft model with different transplantation sites. Results Mouse ovaries treated with DXR in vitro and in vivo had reduced PMF populations and damaged follicle health. We did not observe effect of DXR-induced PMF loss or damage to follicle/stromal health in human ovarian cortex, this might have been due to an insufficient dose or duration of DXR. Although AMH does not prevent DXR-induced PMF loss in pre-pubertal and adult mouse ovaries, in mouse ovaries treated with higher concentration of AMH in vitro, DXR did not cause a significant loss in PMFs. This is the first study to illustrate an effect of AMH on DXR-induced PMF loss on pre-pubertal mouse ovaries. However, more experiments with higher doses of AMH and larger sample size are needed to confirm this finding. Discussion We did not observe that AMH could prevent DXR-induced PMF loss in mouse ovaries in vivo. Further studies are warranted to investigate whether AMH has a protective effect against DXR in xenotransplanted human ovarian tissue. Thus, to obtain robust evidence about the potential of AMH in fertility preservation during chemotherapy treatment, alternative AMH administration strategies need to be explored alongside DXR administration to fully interrogate the effect of DXR and AMH on human xenografted tissues.
Collapse
Affiliation(s)
- Xi Wei
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Briet D Bjarkadottir
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Devi Nadjaja
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Sairah Sheikh
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Muhammad Fatum
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Fertility, Institute of Reproductive Sciences, Oxford, United Kingdom
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Suzannah A Williams
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Zheng X, Liu X, Zhang X, Zhao Z, Wu W, Yu S. A single-cell and spatially resolved atlas of human osteosarcomas. J Hematol Oncol 2024; 17:71. [PMID: 39164791 PMCID: PMC11337771 DOI: 10.1186/s13045-024-01598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/15/2024] [Indexed: 08/22/2024] Open
Abstract
Osteosarcomas are intricate cellular ecosystems, where heterotypic interactions significantly influence disease progression and therapeutic outcomes. Despite their importance, a detailed understanding of their cellular composition and organizational structure remains elusive. In this study, we provide a comprehensive single-cell and spatially resolved transcriptomics analysis of human osteosarcomas. We construct a cellular meta-map to dissect spatial transcriptomic data, unveiling a detailed atlas of osteosarcoma compositional subgroups. We meticulously characterize the unique gene signatures and functional states of each subgroup and investigate the impact of chemotherapy on these cellular subpopulations. Additionally, our spatial transcriptomics analysis identifies a distinct spatial niche, located at the forefront of tumor necrotic zones, potentially associated with chemotherapy resistance. We also delve into the crosstalk between different cellular subgroups. This study furnishes a comprehensive transcriptional atlas of osteosarcoma's cellular architecture, enriching our comprehension of its complexity and laying the groundwork for more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Xuejing Zheng
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xu Liu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinxin Zhang
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhenguo Zhao
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wence Wu
- Department of Orthopedics, Peking University First Hospital, Beijing, 100021, China
| | - Shengji Yu
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
13
|
Shaygani F, Jalali K, Javanmardi Fard H, Afrasiabi Z, Ahmadi Marzaleh M. Exploring the lived experience of mothers of children with leukemia: a qualitative study from Iran. BMC Womens Health 2024; 24:457. [PMID: 39152443 PMCID: PMC11328460 DOI: 10.1186/s12905-024-03300-y] [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: 11/20/2023] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Leukemia, as one of the most common pediatric cancers, has negatively affected many children around the world. Parents often experience increased feeling of distress shortly after being informed about their child's diagnosis. The distress experienced by parents can adversely affect various aspects of their life. This study aimed to develop an understanding of the lived experience of the mothers whose children suffer from leukemia in Shiraz, Iran. METHODS This phenomenological study was performed from April to August 2023, and 10 people were selected as participants by purposive sampling. In-depth and semi-structured interviews were performed for collecting the data. RESULTS The participants' lived experiences during their children's leukemia were classified into five main categories, namely behavioral problems, spiritual issues, psychological problems, issues related to treatment, and economic matters. CONCLUSION Knowing the experiences of parents, especially mothers, in managing and planning for the care of these children seems essential.
Collapse
Affiliation(s)
- Fatemeh Shaygani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Katayoun Jalali
- Department of Medical Education, Clinical Education Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hana Javanmardi Fard
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Afrasiabi
- Midwifery Department, Estahban Branch, Islamic Azad University, Estahban, Iran
| | - Milad Ahmadi Marzaleh
- Department of Health in Disasters and Emergencies, Health Human Resources Research Center, School of Health Management and Information Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Collins M, Gorgoglione R, Impedovo V, Pan X, Chakkarai S, Yi SS, Lodi A, Tiziani S. Exploration of the intracellular chiral metabolome in pediatric BCP-ALL: a pilot study investigating the metabolic phenotype of IgH locus aberrations. Front Oncol 2024; 14:1413264. [PMID: 39161381 PMCID: PMC11332069 DOI: 10.3389/fonc.2024.1413264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/13/2024] [Indexed: 08/21/2024] Open
Abstract
Background and aims Aberrations in the immunoglobulin heavy chain (IgH) locus are associated with poor prognosis in pediatric precursor B-cell acute lymphoblastic leukemia (BCP-ALL) patients. The primary objective of this pilot study is to enhance our understanding of the IgH phenotype by exploring the intracellular chiral metabolome. Materials and methods Leukemia cells were isolated from the bone marrow of BCP-ALL pediatric patients at diagnosis. The samples' metabolome and transcriptome were characterized using untargeted chiral metabolomic and next-generation sequencing transcriptomic analyses. Results For the first time D- amino acids were identified in the leukemic cells' intracellular metabolome from the bone marrow niche. Chiral metabolic signatures at diagnosis was indicative of a resistant phenotype. Through integrated network analysis and Pearson correlation, confirmation was obtained regarding the association of the IgH phenotype with several genes linked to poor prognosis. Conclusion The findings of this study have contributed to the understanding that the chiral metabolome plays a role in the poor prognosis observed in an exceptionally rare patient cohort. The findings include elevated D-amino acid incorporation in the IgH group, the emergence of several unknown, potentially enantiomeric, metabolites, and insights into metabolic pathways that all warrant further exploration.
Collapse
Affiliation(s)
- Meghan Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Ruggiero Gorgoglione
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Valeria Impedovo
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Sathyaseelan Chakkarai
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - S. Stephen Yi
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Interdisciplinary Life Sciences Graduate Programs, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Oden Institute for Computational Engineering and Sciences, and Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, United States
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
15
|
Garg HK, Shashi KK, Fisher P, Winant AJ, Hull NC, Lee EY. Pediatric Upper Abdominal Masses: Current Practical Imaging Assessment. Semin Roentgenol 2024; 59:299-311. [PMID: 38997183 DOI: 10.1053/j.ro.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 07/14/2024]
Affiliation(s)
- Harsha K Garg
- Department of Diagnostic, Molecular and Interventional Radiology and Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY.
| | - Kumar K Shashi
- Department of Radiology, Arkansas Children's Hospital, Little Rock, AR
| | - Paul Fisher
- Department of Diagnostic, Molecular and Interventional Radiology and Pediatrics, Kravis Children's Hospital, Icahn School of Medicine at The Mount Sinai Hospital, New York, NY
| | - Abbey J Winant
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Nathan C Hull
- Department of Radiology, Division of Pediatric Radiology, Mayo Clinic, Rochester, MN
| | - Edward Y Lee
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Hodder A, Leiter SM, Kennedy J, Addy D, Ahmed M, Ajithkumar T, Allinson K, Ancliff P, Bailey S, Barnard G, Burke GAA, Burns C, Cano-Flanagan J, Chalker J, Coleman N, Cheng D, Clinch Y, Dryden C, Ghorashian S, Griffin B, Horan G, Hubank M, May P, McDerra J, Nagrecha R, Nicholson J, O'Connor D, Pavasovic V, Quaegebeur A, Rao A, Roberts T, Samarasinghe S, Stasevich I, Tadross JA, Trayers C, Trotman J, Vora A, Watkins J, Chitty LS, Bowdin S, Armstrong R, Murray MJ, Hook CE, Tarpey P, Vedi A, Bartram J, Behjati S. Benefits for children with suspected cancer from routine whole-genome sequencing. Nat Med 2024; 30:1905-1912. [PMID: 38956197 PMCID: PMC11271414 DOI: 10.1038/s41591-024-03056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/08/2024] [Indexed: 07/04/2024]
Abstract
Clinical whole-genome sequencing (WGS) has been shown to deliver potential benefits to children with cancer and to alter treatment in high-risk patient groups. It remains unknown whether offering WGS to every child with suspected cancer can change patient management. We collected WGS variant calls and clinical and diagnostic information from 281 children (282 tumors) across two English units (n = 152 from a hematology center, n = 130 from a solid tumor center) where WGS had become a routine test. Our key finding was that variants uniquely attributable to WGS changed the management in ~7% (20 out of 282) of cases while providing additional disease-relevant findings, beyond standard-of-care molecular tests, in 108 instances for 83 (29%) cases. Furthermore, WGS faithfully reproduced every standard-of-care molecular test (n = 738) and revealed several previously unknown genomic features of childhood tumors. We show that WGS can be delivered as part of routine clinical care to children with suspected cancer and can change clinical management by delivering unexpected genomic insights. Our experience portrays WGS as a clinically impactful assay for routine practice, providing opportunities for assay consolidation and for delivery of molecularly informed patient care.
Collapse
Affiliation(s)
- Angus Hodder
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Sarah M Leiter
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Jonathan Kennedy
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Dilys Addy
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Munaza Ahmed
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Kieren Allinson
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Phil Ancliff
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Shivani Bailey
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Gemma Barnard
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - G A Amos Burke
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Charlotte Burns
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | - Nicholas Coleman
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Danny Cheng
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Caryl Dryden
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Sara Ghorashian
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Blanche Griffin
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- North Thames Genomic Laboratory Hub, London, UK
| | - Gail Horan
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Michael Hubank
- North Thames Genomic Laboratory Hub, London, UK
- The Institute of Cancer Research, London, UK
| | - Phillippa May
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Joanna McDerra
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Rajvi Nagrecha
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - James Nicholson
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - David O'Connor
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Vesna Pavasovic
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Annelies Quaegebeur
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Anupama Rao
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Thomas Roberts
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | | | - Iryna Stasevich
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - John A Tadross
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
- MRC Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Claire Trayers
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jamie Trotman
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | - Ajay Vora
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - James Watkins
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | - Lyn S Chitty
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- North Thames Genomic Laboratory Hub, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sarah Bowdin
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Genomics Laboratory Hub, Cambridge, UK
| | - Ruth Armstrong
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Matthew J Murray
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Catherine E Hook
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Patrick Tarpey
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- East Genomics Laboratory Hub, Cambridge, UK.
| | - Aditi Vedi
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| | - Jack Bartram
- Great Ormond Street Hospital NHS Foundation Trust, London, UK.
- North Thames Genomic Laboratory Hub, London, UK.
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Zheng Y, Zhang C, Sun X, Kang K, Luo R, Zhao A, Wu Y. Survival trend and outcome prediction for pediatric Hodgkin and non-Hodgkin lymphomas based on machine learning. Clin Exp Med 2024; 24:132. [PMID: 38890203 PMCID: PMC11189314 DOI: 10.1007/s10238-024-01402-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Pediatric Hodgkin and non-Hodgkin lymphomas differ from adult cases in biology and management, yet there is a lack of survival analysis tailored to pediatric lymphoma. We analyzed lymphoma data from 1975 to 2018, comparing survival trends between 7,871 pediatric and 226,211 adult patients, identified key risk factors for pediatric lymphoma survival, developed a predictive nomogram, and utilized machine learning to predict long-term lymphoma-specific mortality risk. Between 1975 and 2018, we observed substantial increases in 1-year (19.3%), 5-year (41.9%), and 10-year (48.8%) overall survival rates in pediatric patients with lymphoma. Prognostic factors such as age, sex, race, Ann Arbor stage, lymphoma subtypes, and radiotherapy were incorporated into the nomogram. The nomogram exhibited excellent predictive performance with area under the curve (AUC) values of 0.766, 0.724, and 0.703 for one-year, five-year, and ten-year survival, respectively, in the training cohort, and AUC values of 0.776, 0.712, and 0.696 in the validation cohort. Importantly, the nomogram outperformed the Ann Arbor staging system in survival prediction. Machine learning models achieved AUC values of approximately 0.75, surpassing the conventional method (AUC = ~ 0.70) in predicting the risk of lymphoma-specific death. We also observed that pediatric lymphoma survivors had a substantially reduced risk of lymphoma after ten years b,ut faced an increasing risk of non-lymphoma diseases. The study highlights substantial improvements in pediatric lymphoma survival, offers reliable predictive tools, and underscores the importance of long-term monitoring for non-lymphoma health issues in pediatric patients.
Collapse
Grants
- No. 82303773, No. 82303772, No. 82204490, No. 82303694 National Natural Science Foundation of China
- No. 82303773, No. 82303772, No. 82204490, No. 82303694 National Natural Science Foundation of China
- No. 82303773, No. 82303772, No. 82204490, No. 82303694 National Natural Science Foundation of China
- No. 82303773, No. 82303772, No. 82204490, No. 82303694 National Natural Science Foundation of China
- No. 2023NSFSC1885 Natural Science Foundation of Sichuan Province
- No. 2023NSFSC1885 Natural Science Foundation of Sichuan Province
- No. 2023YFS0306 Key Research, Development Program of Sichuan Province
- No. 2023YFS0306 Key Research, Development Program of Sichuan Province
- No. GZB20230481 Postdoctoral Fellowship Program of CPSF
- No. 2024HXBH149, No. 2024HXBH006 Post-Doctor Research Project, West China Hospital, Sichuan University
Collapse
Affiliation(s)
- Yue Zheng
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunlan Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Sun
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ren Luo
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.
| | - Yijun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Pathania AS, Chava H, Chaturvedi NK, Chava S, Byrareddy SN, Coulter DW, Challagundla KB. The miR-29 family facilitates the activation of NK-cell immune responses by targeting the B7-H3 immune checkpoint in neuroblastoma. Cell Death Dis 2024; 15:428. [PMID: 38890285 PMCID: PMC11189583 DOI: 10.1038/s41419-024-06791-7] [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: 11/20/2023] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
Neuroblastoma (NB) is a highly aggressive pediatric cancer that originates from immature nerve cells, presenting significant treatment challenges due to therapy resistance. Despite intensive treatment, approximately 50% of high-risk NB cases exhibit therapy resistance or experience relapse, resulting in poor outcomes often associated with tumor immune evasion. B7-H3 is an immune checkpoint protein known to inhibit immune responses. MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation. Our study aims to explore the impact of miRNAs on B7-H3 regulation, the anti-tumor immune response, and tumorigenicity in NB. Analysis of NB patients and patient-derived xenograft tumors revealed a correlation between higher B7-H3 expression and poorer patient survival. Notably, deceased patients exhibited a depletion of miR-29 family members (miR-29a, miR-29b, and miR-29c), which displayed an inverse association with B7-H3 expression in NB patients. Overexpression and knockdown experiments demonstrated that these miRNAs degrade B7-H3 mRNA, resulting in enhanced NK cell activation and cytotoxicity. In vivo, experiments provided further evidence that miR-29 family members reduce tumorigenicity, macrophage infiltration, and microvessel density, promote infiltration and activation of NK cells, and induce tumor cell apoptosis. These findings offer a rationale for developing more effective combination treatments that leverage miRNAs to target B7-H3 in NB patients.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Haritha Chava
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Don W Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kishore B Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- The Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
19
|
Reel SM, Siegel RM, Pillay Smiley N. Pediatric Oncology and Obesity: An Introduction for General Pediatricians. Clin Pediatr (Phila) 2024; 63:589-593. [PMID: 37542412 DOI: 10.1177/00099228231191957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/07/2023]
Abstract
For pediatric patients with cancer, a healthy lifestyle is important for treatment outcomes and beyond. General pediatricians play a major role in the care of these patients, particularly given the improved rates of survival. Pediatric obesity has many negative consequences, but it is an area where primary care providers can make an impact and provide support to childhood cancer survivors. To provide the best quality of care for this population, there must be collaboration between primary care and oncology providers. Additionally, general practioners should feel empowered to offer standard nutrition and physical activity recommendations to all childhood cancer survivors. For pediatric patients who carry a cancer diagnosis, cure is no longer the only goal. Pediatric providers across specialties need to work as a team to improve long-term quality of life for these patients, starting with modifiable healthy habits.
Collapse
Affiliation(s)
| | - Robert M Siegel
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Better Health and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Natasha Pillay Smiley
- Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
20
|
Rademacher MJ, Faber ML, Bone KM, Medin JA, Schloemer NJ. Fate control engagement augments NK cell responses in LV/hu-IL-12 transduced sarcoma. Exp Mol Pathol 2024; 137:104898. [PMID: 38729059 DOI: 10.1016/j.yexmp.2024.104898] [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: 10/04/2023] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
INTRODUCTION NK cells are an untapped resource for cancer therapy. Sarcomas transduced with lentiviruses to express human IL-12 are only cleared in mice bearing mature human NK cells. However, systemic inflammation limits IL-12 utilization. Fate control a.k.a. "suicide mechanisms" regulate unchecked systemic inflammation caused by cellular immunotherapies. Despite increasing utilization, there remains limited data on immune consequences or tumor-directed effects of fate control. OBJECTIVES We sought to engage the mutant thymidylate kinase (mTMPK) metabolic fate control system to regulate systemic inflammation and assess the impact on NK cell effector functions. METHODS Primary human sarcoma short-passage samples and cell lines were transduced with LV/hu-IL-12_mTMPK engineering expression of IL-12 and an AZT-associated fate control enzyme. We assessed transduced sarcoma responses to AZT engagement and subsequent modulation of NK cell functions as measured by inflammatory cytokine production and cytotoxicity. RESULTS AZT administration to transduced (LV/hu-IL-12_mTMPK) short-passage primary human sarcomas and human Ewing sarcoma, osteosarcoma, and rhabdomyosarcoma cell lines, abrogated the robust expression of human IL-12. Fate control activation elicited a specific dose-dependent cytotoxic effect measured by metabolic activity (WST-1) and cell death (Incucyte). NK effector functions of IFN-γ and cytotoxic granule release were significantly augmented despite IL-12 abrogation. This correlated with preferentially induced expression of NK cell activation ligands. CONCLUSIONS mTMPK fate control engagement terminates transduced sarcoma IL-12 production and triggers cell death, but also augments an NK cell-mediated response coinciding with metabolic stress activating surface ligand induction. Fate control engagement could offer a novel immune activation method for NK cell-mediated cancer clearance.
Collapse
Affiliation(s)
- Mary Jo Rademacher
- Departments of Pediatrics; Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary L Faber
- Departments of Pediatrics; Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kathleen M Bone
- Departments of Pathology; Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jeffrey A Medin
- Departments of Pediatrics; Medical College of Wisconsin, Milwaukee, WI 53226, USA; Departments of Biochemisty; Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Nathan J Schloemer
- Departments of Pediatrics; Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
21
|
Chan C, Stip M, Nederend M, Jansen M, Passchier E, van den Ham F, Wienke J, van Tetering G, Leusen J. Enhancing IgA-mediated neutrophil cytotoxicity against neuroblastoma by CD47 blockade. J Immunother Cancer 2024; 12:e008478. [PMID: 38782540 PMCID: PMC11116899 DOI: 10.1136/jitc-2023-008478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Approximately half of the neuroblastoma patients develop high-risk neuroblastoma. Current treatment involves a multimodal strategy, including immunotherapy with dinutuximab (IgG ch14.18) targeting GD2. Despite achieving promising results, the recurrence rate remains high and poor survival persists. The therapeutic efficacy of dinutuximab is compromised by suboptimal activation of neutrophils and severe neuropathic pain, partially induced by complement activation. METHODS To enhance neutrophil cytotoxicity, IgG ch14.18 was converted to the IgA isotype, resulting in potent neutrophil-mediated antibody-dependent cell-mediated cytotoxicity (ADCC), without complement activation. However, myeloid checkpoint molecules hamper neutrophil cytotoxicity, for example through CD47 that is overexpressed on neuroblastomas and orchestrates an immunosuppressive environment upon ligation to signal regulatory protein alpha (SIRPα) expressed on neutrophils. In this study, we combined IgA therapy with CD47 blockade. RESULTS In vitro killing assays showed enhanced IgA-mediated ADCC by neutrophils targeting neuroblastoma cell lines and organoids in comparison to IgG. Notably, when combined with CD47 blockade, both IgG and IgA therapy were enhanced, though the combination with IgA resulted in the greatest improvement of ADCC. Furthermore, in a neuroblastoma xenograft model, we systemically blocked CD47 with a SIRPα fusion protein containing an ablated IgG1 Fc, and compared IgA therapy to IgG therapy. Only IgA therapy combined with CD47 blockade increased neutrophil influx to the tumor microenvironment. Moreover, the IgA combination strategy hampered tumor outgrowth most effectively and prolonged tumor-specific survival. CONCLUSION These promising results highlight the potential to enhance immunotherapy efficacy against high-risk neuroblastoma through improved neutrophil cytotoxicity by combining IgA therapy with CD47 blockade.
Collapse
Affiliation(s)
- Chilam Chan
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Marjolein Stip
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Maaike Nederend
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Marco Jansen
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | | | - Femke van den Ham
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Judith Wienke
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Geert van Tetering
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Jeanette Leusen
- Center for Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
22
|
Deyell RJ, Shen Y, Titmuss E, Dixon K, Williamson LM, Pleasance E, Nelson JMT, Abbasi S, Krzywinski M, Armstrong L, Bonakdar M, Ch'ng C, Chuah E, Dunham C, Fok A, Jones M, Lee AF, Ma Y, Moore RA, Mungall AJ, Mungall KL, Rogers PC, Schrader KA, Virani A, Wee K, Young SS, Zhao Y, Jones SJM, Laskin J, Marra MA, Rassekh SR. Whole genome and transcriptome integrated analyses guide clinical care of pediatric poor prognosis cancers. Nat Commun 2024; 15:4165. [PMID: 38755180 PMCID: PMC11099106 DOI: 10.1038/s41467-024-48363-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
The role for routine whole genome and transcriptome analysis (WGTA) for poor prognosis pediatric cancers remains undetermined. Here, we characterize somatic mutations, structural rearrangements, copy number variants, gene expression, immuno-profiles and germline cancer predisposition variants in children and adolescents with relapsed, refractory or poor prognosis malignancies who underwent somatic WGTA and matched germline sequencing. Seventy-nine participants with a median age at enrollment of 8.8 y (range 6 months to 21.2 y) are included. Germline pathogenic/likely pathogenic variants are identified in 12% of participants, of which 60% were not known prior. Therapeutically actionable variants are identified by targeted gene report and whole genome in 32% and 62% of participants, respectively, and increase to 96% after integrating transcriptome analyses. Thirty-two molecularly informed therapies are pursued in 28 participants with 54% achieving a clinical benefit rate; objective response or stable disease ≥6 months. Integrated WGTA identifies therapeutically actionable variants in almost all tumors and are directly translatable to clinical care of children with poor prognosis cancers.
Collapse
Affiliation(s)
- Rebecca J Deyell
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada.
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Emma Titmuss
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Katherine Dixon
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Jessica M T Nelson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Sanna Abbasi
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Martin Krzywinski
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Linlea Armstrong
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Melika Bonakdar
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Carolyn Ch'ng
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Chris Dunham
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Alexandra Fok
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Martin Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Anna F Lee
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yussanne Ma
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Paul C Rogers
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada
| | - Kasmintan A Schrader
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Alice Virani
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Kathleen Wee
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Sean S Young
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Cancer Genetics and Genomics Laboratory, Department of Pathology and Laboratory Medicine, BC Cancer, Vancouver, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Shahrad R Rassekh
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
23
|
Wyatt KD, Birz S, Castellino SM, Henderson TO, Lucas JT, Pei Q, Zhou Y, Volchenboum SL, Furner B, Watkins M, Kelly KM, Flerlage JE. Accelerating pediatric Hodgkin lymphoma research: the Hodgkin Lymphoma Data Collaboration (NODAL). J Natl Cancer Inst 2024; 116:642-646. [PMID: 38273668 PMCID: PMC11077302 DOI: 10.1093/jnci/djae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
Data commons have proven to be an indispensable avenue for advancing pediatric cancer research by serving as unified information technology platforms that, when coupled with data standards, facilitate data sharing. The Pediatric Cancer Data Commons, the flagship project of Data for the Common Good (D4CG), collaborates with disease-based consortia to facilitate development of clinical data standards, harmonization and pooling of clinical data from disparate sources, establishment of governance structure, and sharing of clinical data. In the interest of international collaboration, researchers developed the Hodgkin Lymphoma Data Collaboration and forged a relationship with the Pediatric Cancer Data Commons to establish a data commons for pediatric Hodgkin lymphoma. Herein, we describe the progress made in the formation of Hodgkin Lymphoma Data Collaboration and foundational goals to advance pediatric Hodgkin lymphoma research.
Collapse
Affiliation(s)
- Kirk D Wyatt
- Department of Pediatric Hematology/Oncology, Roger Maris Cancer Center, Sanford Health, Fargo, ND, USA
| | - Suzi Birz
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Sharon M Castellino
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Cancer and Blood Disorders Center, Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Tara O Henderson
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - John T Lucas
- Department of Radiation Oncology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Qinglin Pei
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Yiwang Zhou
- Department of Biostatistics, St Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Brian Furner
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Michael Watkins
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Kara M Kelly
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Jamie E Flerlage
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
24
|
Kappelmann L, Götte M, Krombholz A, Hüter J, Fischer B. Factors That Influence Physical Activity Behavior in Children and Adolescents During and After Cancer Treatment: A Qualitative Systematic Review of the Literature. Pediatr Exerc Sci 2024; 36:106-114. [PMID: 37890837 DOI: 10.1123/pes.2022-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 07/31/2023] [Accepted: 08/23/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE The aim of this systematic review is to reveal the social, personal, and contextual factors that influence physical activity (PA) in children and adolescents during and after cancer treatment. METHOD SPORTDiscus, Cochrane, Web of Science, PubMed, and FIS Education electronic database were systematically searched. RESULTS The 13 included studies show that social support (parents, siblings, and friends) in particular is rated as important by cancer survivors; for example, doing PA together. Depending on the treatment status and state of health, particularities arise. During the acute treatment phase, parents issued more prohibitions regarding PA than after treatment. The state of health and concern about infections are described as inhibiting factors. Not all hospitals generally offer special exercise programs for cancer patients, and in some cases, only sporadic exercise sessions were conducted by specialized staff. In addition, the hospital atmosphere, such as cramped rooms, tends to be associated with demotivating effects. CONCLUSIONS Both inhibiting and promoting factors in the area of social, personal, and contextual factors could be identified. The most fundamental factor for PA is the physical condition. Social factors, such as parents or friends, often have a motivating effect and can promote PA. Inhibiting factors are mainly context-related, such as an environment unsuitable for PA. Although the review highlights interesting aspects, further treatment-related and longitudinal studies could provide deeper insights.
Collapse
Affiliation(s)
- Laura Kappelmann
- Department of Sport Science, Christian-Albrechts-University of Kiel, Kiel,Germany
| | - Miriam Götte
- West German Cancer Center, University Hospital Essen, Essen,Germany
| | - Arno Krombholz
- Faculty of Sport Science, Ruhr-University Bochum, Bochum,Germany
| | - Jan Hüter
- Faculty of Sport Science, Ruhr-University Bochum, Bochum,Germany
| | - Britta Fischer
- Department of Sport Science, Christian-Albrechts-University of Kiel, Kiel,Germany
| |
Collapse
|
25
|
Liu W, Cheng H, Huang Z, Li Y, Zhang Y, Yang Y, Jin T, Sun Y, Deng Z, Zhang Q, Lou F, Cao S, Wang H, Niu X. The correlation between clinical outcomes and genomic analysis with high risk factors for the progression of osteosarcoma. Mol Oncol 2024; 18:939-955. [PMID: 37727135 PMCID: PMC10994228 DOI: 10.1002/1878-0261.13526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 09/21/2023] Open
Abstract
Osteosarcoma (OS) is a rare but aggressive malignancy. Despite previous reports, molecular characterization of this disease is not well understood, and little is known regarding OS in Chinese patients. Herein, we analyzed the genomic signatures of 73 Chinese OS cases. TP53, NCOR1, LRP1B, ATRX, RB1, and TFE3 were the most frequently mutated gene in our OS cohort. In addition, the genomic analysis of Western OS patients was performed. Notably, there were remarkable disparities in mutational landscape, base substitution pattern, and tumor mutational burden between the Chinese and Western OS cohorts. Specific molecular mechanisms, including DNA damage repair (DDR) gene mutations, copy number variation (CNV) presence, aneuploidy, and intratumoral heterogeneity, were associated with disease progression. Additionally, 30.1% of OS patients carried clinically actionable alterations, which were mainly enriched in PI3K, MAPK, DDR, and RTK signaling pathways. A specific molecular subtype incorporating DDR alterations and CNVs was significantly correlated with distant metastasis-free survival and event-free survival, and this correlation was observed in all subgroups with different characteristics. These findings comprehensively elucidated the genomic profile and revealed novel prognostic factors in OS, which would contribute to understanding this disease and promoting precision medicine of this population.
Collapse
Affiliation(s)
- Weifeng Liu
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | | | - Zhen Huang
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | - Yaping Li
- Acornmed Biotechnology Co., Ltd.BeijingChina
| | | | - Yongkun Yang
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | - Tao Jin
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | - Yang Sun
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | - Zhiping Deng
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | - Qing Zhang
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| | - Feng Lou
- Acornmed Biotechnology Co., Ltd.BeijingChina
| | - Shanbo Cao
- Acornmed Biotechnology Co., Ltd.BeijingChina
| | - Huina Wang
- Acornmed Biotechnology Co., Ltd.BeijingChina
| | - Xiaohui Niu
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan HospitalPeking UniversityBeijingChina
- Fourth Medical College of Peking UniversityBeijingChina
- National Center for OrthopeadicsBeijingChina
| |
Collapse
|
26
|
Rashidian P. An update on oncofertility in prepubertal females. J Gynecol Obstet Hum Reprod 2024; 53:102742. [PMID: 38341083 DOI: 10.1016/j.jogoh.2024.102742] [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: 09/25/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
Cancer is a life-threatening event for pediatric patients. Treatment advancements in pediatric cancer have improved prognosis, but some of these treatments have gonadotoxic potential and may affect fertility in different ways. Due to the growing interest of the research community in the life prospects of young cancer survivors, there has been a demand to intersect reproductive medicine and oncology, which is referred to as "oncofertility". There are various fertility preservation options according to gender and pubertal status, and shared decisions must take place at the time of diagnosis. This study aims to provide a critical review of current and emerging strategies for preserving and restoring fertility in prepubertal females, ranging from established methods to experimental approaches that can be offered before, during, and after anticancer therapies. Additionally, the author aims to review how clinicians' awareness of oncofertility options and the latest advancements in this field, timely referral, and proper consultations with patients and their families are vital in addressing their concerns, providing emotional support, and guiding them through the decision-making process, as well as potential barriers that may hinder the fertility preservation process.
Collapse
Affiliation(s)
- Pegah Rashidian
- Reproductive Health Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
27
|
Thrope A, Gerber LM, Thomas C, Antal Z. Longitudinal assessment of Leydig cell function in male survivors of childhood cancer. Pediatr Blood Cancer 2024; 71:e30829. [PMID: 38149843 PMCID: PMC10843544 DOI: 10.1002/pbc.30829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND As the number and longevity of childhood cancer survivors increases, assessing treatment-associated late effects remains crucial. We longitudinally examined the incidence of and associated risk factors for Leydig cell dysfunction (LCD) and Leydig cell failure (LCF) in men treated for pediatric cancers at our institution. PROCEDURE We performed a retrospective longitudinal cohort study of adult male survivors treated for various childhood cancers who are at risk for LCD. The outcomes of interest were serum testosterone and luteinizing hormone (LH) levels during childhood and adulthood. Risk factors assessed included treatment with stem cell transplant, total body irradiation (TBI), and exposure to alkylating agents. RESULTS Out of 118 eligible subjects, 7.6% had LCF and 14.4% had LCD. Median age at last testosterone level was 20 years. Subjects with sufficient testosterone levels in adulthood (N = 105) remained sufficient for a mean of 11.1 years following completion of cancer treatment. We found significant associations between LCF and treatment with TBI (p < .003) and between LCF in adulthood and testosterone insufficiency in childhood (p < .001). No statistically significant association was found between LCF and cyclophosphamide equivalent dose greater than 20 g/m2 (p = .2). LCF/LCD occurred in a small number of nonirradiated patients treated with the highest doses of alkylators. CONCLUSIONS Incidence of LCF and LCD are low in male survivors of childhood cancer. Longitudinally, there is an association between childhood testosterone insufficiency and LCF in adulthood. Alkylating agents and stem cell transplant without TBI were not associated with LCF in our study.
Collapse
Affiliation(s)
- Alexandra Thrope
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center New York, NY 10065
| | | | | | - Zoltan Antal
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center New York, NY 10065
| |
Collapse
|
28
|
Su Q, Wang N, Wang B, Wang Y, Dai Z, Zhao X, Li X, Li Q, Yang G, Nie P. Ct-based intratumoral and peritumoral radiomics for predicting prognosis in osteosarcoma: A multicenter study. Eur J Radiol 2024; 172:111350. [PMID: 38309216 DOI: 10.1016/j.ejrad.2024.111350] [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: 08/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
PURPOSE To evaluate the performance of CT-based intratumoral, peritumoral and combined radiomics signatures in predicting prognosis in patients with osteosarcoma. METHODS The data of 202 patients (training cohort:102, testing cohort:100) with osteosarcoma admitted to the two hospitals from August 2008 to February 2022 were retrospectively analyzed. Progression free survival (PFS) and overall survival (OS) were used as the end points. The radiomics features were extracted from CT images, three radiomics signatures(RSintratumoral, RSperitumoral, RScombined)were constructed based on intratumoral, peritumoral and combined radiomics features, respectively, and the radiomics score (Rad-score) were calculated. Kaplan-Meier survival analysis was used to evaluate the relationship between the Rad-score with PFS and OS, the Harrell's concordance index (C-index) was used to evaluate the predictive performance of the radiomics signatures. RESULTS Finally, 8, 6, and 21 features were selected for the establishment of RSintratumoral, RSperitumoral, and RScombined, respectively. Kaplan-Meier survival analysis confirmed that the Rad-scores of the three RSs were significantly correlated with the PFS and OS of patients with osteosarcoma. Among the three radiomics signatures, RScombined had better predictive performance, the C-index of PSF prediction was 0.833 in the training cohort and 0.814 in the testing cohort, the C-index of OS prediction was 0.796 in the training cohort and 0.764 in the testing cohort. CONCLUSIONS CT-based intratumoral, peritumoral and combined radiomics signatures can predict the prognosis of patients with osteosarcoma, which may assist in individualized treatment and improving the prognosis of osteosarcoma patients.
Collapse
Affiliation(s)
- Qiushi Su
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ning Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bingyan Wang
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | | | - Zhengjun Dai
- Scientific Research Department, Huiying Medical Technology Co., Ltd, Beijing, China
| | - Xia Zhao
- Department of Radiology, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoli Li
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qiyuan Li
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guangjie Yang
- Department of Nuclear Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Pei Nie
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
29
|
Annetta MG, Celentano D, Zumstein L, Attinà G, Ruggiero A, Conti G, Pittiruti M. Catheter-related complications in onco-hematologic children: A retrospective clinical study on 227 central venous access devices. J Vasc Access 2024; 25:512-518. [PMID: 36113076 DOI: 10.1177/11297298221122128] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND The use of central venous access devices (CVADs) is of paramount importance to safely deliver antiblastic and support therapies in children with cancer. Though, in pediatric patients, as much as in adults, CVADs are potentially associated with severe complications which may result in unscheduled interruption of therapy, hospitalization, increased morbidity/mortality, and increased cost of care. METHODS We have reviewed retrospectively our experience with CVADs in children with solid tumors and hematologic diseases, with the purpose of verifying if the adoption of well-defined insertion and maintenance bundles might be effective in reducing catheter-related complications, and in particular catheter-related thrombosis. RESULTS A total of 227 CVADs were analyzed: 175 peripherally inserted central catheters (PICCs), 50 centrally inserted central catheters (CICCs), and 2 femorally inserted central catheters. All CVADs were non-valved, non-cuffed power injectable polyurethane catheters; 81% were tunneled. Median dwelling time of CVADs was 172 days, for a total number of 39,044 catheter days. A very low incidence of both symptomatic catheter-related thrombosis (0.9%) and catheter-related blood stream infection (0.56 episodes per 1000 catheter days) was found. Unscheduled removal or guidewire replacement because of mechanic complications occurred in 15.7% of CVADs. There was no difference in terms of complications between PICCs and CICCs or between tunneled and non-tunneled catheters. CONCLUSIONS Our experience with CVADs in oncologic and hematologic children suggests that catheter-related complications may be minimized by the adoption of appropriate insertion and maintenance bundles.
Collapse
Affiliation(s)
- Maria Giuseppina Annetta
- Department of Anesthesia and Intensive Care, Catholic University Hospital "A.Gemelli," Rome, Italy
| | - Davide Celentano
- Department of Oncology, Catholic University Hospital "A.Gemelli," Rome, Italy
| | - Lucrezia Zumstein
- Department of Pediatric Oncology, Catholic University Hospital "A.Gemelli," Rome, Italy
| | - Giorgio Attinà
- Department of Pediatric Oncology, Catholic University Hospital "A.Gemelli," Rome, Italy
| | - Antonio Ruggiero
- Department of Pediatric Oncology, Catholic University Hospital "A.Gemelli," Rome, Italy
| | - Giorgio Conti
- Department of Anesthesia and Intensive Care, Catholic University Hospital "A.Gemelli," Rome, Italy
| | - Mauro Pittiruti
- Department of Surgery, Catholic University Hospital "A.Gemelli," Rome, Italy
| |
Collapse
|
30
|
Żebrowska U, Balwierz W, Wechowski J, Wieczorek A. Survival Benefit of Myeloablative Therapy with Autologous Stem Cell Transplantation in High-Risk Neuroblastoma: A Systematic Literature Review. Target Oncol 2024; 19:143-159. [PMID: 38401028 DOI: 10.1007/s11523-024-01033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Multimodal treatment of newly diagnosed high-risk neuroblastoma (HRNB) includes induction chemotherapy, consolidation with myeloablative therapy (MAT) and autologous stem cell transplantation (ASCT), followed by anti-disialoganglioside 2 (GD2) immunotherapy, as recommended by the Children's Oncology Group (COG) and the Society of Paediatric Oncology European Neuroblastoma (SIOPEN). Some centres proposed an alternative approach with induction chemotherapy followed by anti-GD2 immunotherapy, without MAT+ASCT. OBJECTIVE The aim of this systematic literature review was to compare survival outcomes in patients with HRNB treated with or without MAT+ASCT and with or without subsequent anti-GD2 immunotherapy. PATIENTS AND METHODS The review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. MEDLINE via PubMed and EMBASE databases were systematically searched for randomised controlled trials (RCT) and observational comparative studies in patients with HRNB using search terms for 'neuroblastoma' and ('myeloablative therapy' OR 'stem cell transplantation'). Reporting of at least one survival outcome [event-free survival (EFS), progression-free survival, relapse-free survival and/or overall survival (OS)] was required for inclusion. Outcomes from RCTs were synthesized in meta-analysis, while meta-analysis of non-RCTs was not planned owing to expected heterogeneity. RESULTS Literature searches produced 2587 results with 41 publications reporting 34 comparative studies included in the review. Of these, 7 publications reported 4 RCTs, and 34 publications reported 30 non-RCT studies. Studies differed with respect to included populations, induction regimen, response to induction, additional treatments and transplantation procedures. Subsequent treatments of relapse were rarely reported and could not be compared. In the meta-analysis, EFS was in favour of MAT+ASCT over conventional chemotherapy or no further treatment [hazard ratio (HR) = 0.78, 95% confidence interval (CI) 0.67-0.91, p = 0.001] with a trend favouring MAT+ASCT for OS (HR = 0.86, 95% CI 0.73-1.00, p = 0.05). Tandem MAT+ASCT was found to improve EFS compared with the single procedure, with improvement in both EFS and OS in patients treated with anti-GD2 therapy. Non-RCT comparative studies were broadly consistent with evidence from the RCTs; however, not all reported survival benefits of MAT+ASCT (single or tandem). Limited comparative evidence on treatment without MAT+ASCT in patients treated with anti-GD2 immunotherapy suggests an increased risk of relapse. In relapsed patients, MAT+ASCT appears to improve OS, but evidence remains scarce. CONCLUSIONS Survival benefits in patients treated with MAT+ASCT confirm that the procedure should remain an integral part of multimodal therapy. In patients treated with anti-GD2 immunotherapy, limited evidence suggests that omitting MAT+ASCT is associated with an increased risk of relapse, and therefore, a change in clinical practice can currently not be recommended. Evidence suggests the use of tandem MAT+ASCT compared with the single procedure, with greater benefits observed in patients treated with anti-GD2 immunotherapy. Limited evidence also suggests improved survival following MAT+ASCT in relapsed patients, which needs to be viewed in light of emerging chemoimmunotherapy in this setting.
Collapse
Affiliation(s)
- Urszula Żebrowska
- Department of Paediatric Oncology and Haematology, University Children's Hospital of Krakow, 265 Wielicka str, 30-663, Krakow, Poland
| | - Walentyna Balwierz
- Department of Paediatric Oncology and Haematology, University Children's Hospital of Krakow, 265 Wielicka str, 30-663, Krakow, Poland
- Department of Paediatric Oncology and Haematology, Jagiellonian University Medical College, 265 Wielicka str, 30-663, Krakow, Poland
| | - Jarosław Wechowski
- EUSA Pharma, Breakspear Park, Breakspear Way, Hemel Hempstead, HP2 4TZ, UK
| | - Aleksandra Wieczorek
- Department of Paediatric Oncology and Haematology, University Children's Hospital of Krakow, 265 Wielicka str, 30-663, Krakow, Poland.
- Department of Paediatric Oncology and Haematology, Jagiellonian University Medical College, 265 Wielicka str, 30-663, Krakow, Poland.
| |
Collapse
|
31
|
Sarkar S, Stitzlein LM, Chandra J. Impact of electronic cigarettes on pediatric, adolescent and young adult leukemia patients. PEDIATRIC MEDICINE (HONG KONG, CHINA) 2024; 7:3. [PMID: 38616981 PMCID: PMC11014418 DOI: 10.21037/pm-23-43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Electronic cigarettes, which deliver an aerosolized, nicotine-containing product upon inhalation, are a public health issue that continue to gain popularity among adolescents and young adults in the United States. Use of electronic cigarettes is wide, and extends to pediatric patients with multiple comorbidities, including childhood cancer, leaving them vulnerable to further negative health outcomes. Acute leukemias are the most common type of cancer in pediatric populations, and treatment outcomes for these patients are improving; consequently, there is an increased emphasis on the effect of behavioral lifestyle factors on quality of life in survivorship. The rate of electronic cigarette use is higher among pediatric patients with a history of cancer than those without a history of cancer. Because electronic cigarettes are relatively new, much about their acute and long-term consequences remains unknown, as is their effect on therapy outcomes and long-term survivorship. This review article summarizes current knowledge about electronic cigarettes, including their composition and the trends in use among pediatric patients. Furthermore, this review provides a comprehensive description of the impact electronic cigarettes have on leukemia development, treatment and survivorship and highlights gaps in knowledge that will be necessary for developing recommendations, management strategies, and tailored treatments for pediatric leukemia patients and survivors who use these nicotine products.
Collapse
Affiliation(s)
- Sanila Sarkar
- Department of Pediatrics Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lea M. Stitzlein
- Department of Pediatrics Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Joya Chandra
- Department of Pediatrics Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Energy Balance in Cancer Prevention and Survivorship, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Liu Y, Hamid N, Manzoor R, Zhang BF, Liao YL, Wang JX, Pei DS. PPARβ/δ-ANGPTL4 axis mediates the promotion of mono-2-ethylhexyl phthalic acid on MYCN-amplified neuroblastoma development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168949. [PMID: 38042186 DOI: 10.1016/j.scitotenv.2023.168949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/25/2023] [Accepted: 11/25/2023] [Indexed: 12/04/2023]
Abstract
Di-2-ethylhexyl phthalic acid (DEHP) is one of the most widely used plasticizers in the industry, which can improve the flexibility and durability of plastics. It is prone to migrate from various daily plastic products through wear and leaching into the surrounding environment and decompose into the more toxic metabolite mono-2-ethylhexyl phthalic acid (MEHP) after entering the human body. However, the impacts and mechanisms of MEHP on neuroblastoma are unclear. We exposed MYCN-amplified neuroblastoma SK-N-BE(2)C cells to an environmentally related concentration of MEHP and found that MEHP increased the proliferation and migration ability of tumor cells. The peroxisome proliferator-activated receptor (PPAR) β/δ pathway was identified as a pivotal signaling pathway in neuroblastoma, mediating the effects of MEHP through transcriptional sequencing analysis. Because MEHP can bind to the PPARβ/δ protein and initiate the expression of the downstream gene angiopoietin-like 4 (ANGPTL4), the PPARβ/δ-specific agonist GW501516 and antagonist GSK3787, the recombinant human ANGPTL4 protein, and the knockdown of gene expression confirmed the regulation of the PPARβ/δ-ANGPTL4 axis on the malignant phenotype of neuroblastoma. Based on the critical role of PPARβ/δ and ANGPTL4 in the metabolic process, a non-targeted metabolomics analysis revealed that MEHP altered multiple metabolic pathways, particularly lipid metabolites involving fatty acyls, glycerophospholipids, and sterol lipids, which may also be potential factors promoting tumor progression. We have demonstrated for the first time that MEHP can target binding to PPARβ/δ and affect the progression of neuroblastoma by activating the PPARβ/δ-ANGPTL4 axis. This mechanism confirms the health risks of plasticizers as tumor promoters and provides new data support for targeted prevention and treatment of neuroblastoma.
Collapse
Affiliation(s)
- Yiyun Liu
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Naima Hamid
- Faculty of Science and Marine Environment, Ocean Pollution and Ecotoxicology (OPEC) Research Group, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Rakia Manzoor
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Bao-Fu Zhang
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yan-Ling Liao
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jin-Xia Wang
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
33
|
Wienke J, Visser LL, Kholosy WM, Keller KM, Barisa M, Poon E, Munnings-Tomes S, Himsworth C, Calton E, Rodriguez A, Bernardi R, van den Ham F, van Hooff SR, Matser YAH, Tas ML, Langenberg KPS, Lijnzaad P, Borst AL, Zappa E, Bergsma FJ, Strijker JGM, Verhoeven BM, Mei S, Kramdi A, Restuadi R, Sanchez-Bernabeu A, Cornel AM, Holstege FCP, Gray JC, Tytgat GAM, Scheijde-Vermeulen MA, Wijnen MHWA, Dierselhuis MP, Straathof K, Behjati S, Wu W, Heck AJR, Koster J, Nierkens S, Janoueix-Lerosey I, de Krijger RR, Baryawno N, Chesler L, Anderson J, Caron HN, Margaritis T, van Noesel MM, Molenaar JJ. Integrative analysis of neuroblastoma by single-cell RNA sequencing identifies the NECTIN2-TIGIT axis as a target for immunotherapy. Cancer Cell 2024; 42:283-300.e8. [PMID: 38181797 PMCID: PMC10864003 DOI: 10.1016/j.ccell.2023.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 11/10/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024]
Abstract
Pediatric patients with high-risk neuroblastoma have poor survival rates and urgently need more effective treatment options with less side effects. Since novel and improved immunotherapies may fill this need, we dissect the immunoregulatory interactions in neuroblastoma by single-cell RNA-sequencing of 24 tumors (10 pre- and 14 post-chemotherapy, including 5 pairs) to identify strategies for optimizing immunotherapy efficacy. Neuroblastomas are infiltrated by natural killer (NK), T and B cells, and immunosuppressive myeloid populations. NK cells show reduced cytotoxicity and T cells have a dysfunctional profile. Interaction analysis reveals a vast immunoregulatory network and identifies NECTIN2-TIGIT as a crucial immune checkpoint. Combined blockade of TIGIT and PD-L1 significantly reduces neuroblastoma growth, with complete responses (CR) in vivo. Moreover, addition of TIGIT+PD-L1 blockade to standard relapse treatment in a chemotherapy-resistant Th-ALKF1174L/MYCN 129/SvJ syngeneic model induces CR. In conclusion, our integrative analysis provides promising targets and a rationale for immunotherapeutic combination strategies.
Collapse
Affiliation(s)
- Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Lindy L Visser
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Waleed M Kholosy
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kaylee M Keller
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Evon Poon
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Sophie Munnings-Tomes
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Courtney Himsworth
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Elizabeth Calton
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | | | - Ronald Bernardi
- Genentech, A Member of the Roche Group, South San Francisco, CA, USA
| | - Femke van den Ham
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Yvette A H Matser
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Michelle L Tas
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Philip Lijnzaad
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Anne L Borst
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Elisa Zappa
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | - Bronte M Verhoeven
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Shenglin Mei
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Amira Kramdi
- Institut Curie, Inserm U830, PSL Research University, Diversity and Plasticity of Childhood Tumors Lab, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Restuadi Restuadi
- Infection, Immunity and Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK; NIHR Biomedical Research Centre, Great Ormond Street Hospital, London, UK
| | - Alvaro Sanchez-Bernabeu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht University, Utrecht, the Netherlands
| | - Annelisa M Cornel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Juliet C Gray
- Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | | | | | - Marc H W A Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Karin Straathof
- University College London (UCL) Great Ormond Street Institute of Child Health, London, UK; UCL Cancer Institute, London, UK
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht University, Utrecht, the Netherlands; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Netherlands Proteomics Centre, Utrecht University, Utrecht, the Netherlands
| | - Jan Koster
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Isabelle Janoueix-Lerosey
- Institut Curie, Inserm U830, PSL Research University, Diversity and Plasticity of Childhood Tumors Lab, Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, England, UK
| | | | | | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Division Imaging & Cancer, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
34
|
Wang Y, Ma X, Xu E, Huang Z, Yang C, Zhu K, Dong Y, Zhang C. Identifying squalene epoxidase as a metabolic vulnerability in high-risk osteosarcoma using an artificial intelligence-derived prognostic index. Clin Transl Med 2024; 14:e1586. [PMID: 38372422 PMCID: PMC10875711 DOI: 10.1002/ctm2.1586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Osteosarcoma (OSA) presents a clinical challenge and has a low 5-year survival rate. Currently, the lack of advanced stratification models makes personalized therapy difficult. This study aims to identify novel biomarkers to stratify high-risk OSA patients and guide treatment. METHODS We combined 10 machine-learning algorithms into 101 combinations, from which the optimal model was established for predicting overall survival based on transcriptomic profiles for 254 samples. Alterations in transcriptomic, genomic and epigenomic landscapes were assessed to elucidate mechanisms driving poor prognosis. Single-cell RNA sequencing (scRNA-seq) unveiled genes overexpressed in OSA cells as potential therapeutic targets, one of which was validated via tissue staining, knockdown and pharmacological inhibition. We characterized changes in multiple phenotypes, including proliferation, colony formation, migration, invasion, apoptosis, chemosensitivity and in vivo tumourigenicity. RNA-seq and Western blotting elucidated the impact of squalene epoxidase (SQLE) suppression on signalling pathways. RESULTS The artificial intelligence-derived prognostic index (AIDPI), generated by our model, was an independent prognostic biomarker, outperforming clinicopathological factors and previously published signatures. Incorporating the AIDPI with clinical factors into a nomogram improved predictive accuracy. For user convenience, both the model and nomogram are accessible online. Patients in the high-AIDPI group exhibited chemoresistance, coupled with overexpression of MYC and SQLE, increased mTORC1 signalling, disrupted PI3K-Akt signalling, and diminished immune infiltration. ScRNA-seq revealed high expression of MYC and SQLE in OSA cells. Elevated SQLE expression correlated with chemoresistance and worse outcomes in OSA patients. Therapeutically, silencing SQLE suppressed OSA malignancy and enhanced chemosensitivity, mediated by cholesterol depletion and suppression of the FAK/PI3K/Akt/mTOR pathway. Furthermore, the SQLE-specific inhibitor FR194738 demonstrated anti-OSA effects in vivo and exhibited synergistic effects with chemotherapeutic agents. CONCLUSIONS AIDPI is a robust biomarker for identifying the high-risk subset of OSA patients. The SQLE protein emerges as a metabolic vulnerability in these patients, providing a target with translational potential.
Collapse
Affiliation(s)
- Yongjie Wang
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
- Proteomics and Cancer Cell Signaling Group, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Xiaolong Ma
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Enjie Xu
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Zhen Huang
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Kunpeng Zhu
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Yang Dong
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong UniversityShanghaiP. R. China
| | - Chunlin Zhang
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| |
Collapse
|
35
|
Helms L, Guimera AE, Janeway KA, Bailey KM. Innovations in Cancer Treatment of Children. Pediatrics 2023; 152:e2023061539. [PMID: 37920939 PMCID: PMC10657776 DOI: 10.1542/peds.2023-061539] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 11/04/2023] Open
Abstract
Pediatric cancer outcomes have significantly improved, and yet this success is not spread equally across cancer types or patients. Disparities data in pediatric oncology highlight needed improvements in access to care, including clinical trials and advanced testing for all patients. For cancers such as brain tumors and sarcomas, continued advancement in understanding the biology of tumor heterogeneity is an essential step toward finding new therapeutic combinations to improve outcomes. Pediatric cancer survivors need access to emerging technologies aimed at reducing or better managing toxicities from therapy. With advances in treatment and survival, pediatric oncology patients continue to need longitudinal, multidisciplinary subspecialty care. Refining the communication between pediatric oncologists, primary pediatricians, survivorship clinics, and adult primary care is key in ensuring the best lifelong care of pediatric cancer survivors. In this State-of-The-Art review, we discuss 5 major domains in pediatric oncology: reducing toxicity, cancer biology, novel therapies, detection and monitoring, and access to care, to highlight recent advances and areas for continued improvement.
Collapse
Affiliation(s)
- Lauren Helms
- Department of Pediatrics, Michigan Medicine, Ann Arbor, Michigan
| | - Allison E. Guimera
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California
| | - Katherine A. Janeway
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Kelly M. Bailey
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Rodrigues WF, Miguel CB, de Abreu MCM, Neto JM, Oliveira CJF. Potential Associations between Vascular Biology and Hodgkin's Lymphoma: An Overview. Cancers (Basel) 2023; 15:5299. [PMID: 37958472 PMCID: PMC10649902 DOI: 10.3390/cancers15215299] [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: 08/24/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Hodgkin's lymphoma (HL) is a lymphatic neoplasm typically found in the cervical lymph nodes. The disease is multifactorial, and in recent years, the relationships between various vascular molecules have been explored in the field of vascular biology. The connection between vascular biology and HL is intricate and the roles of several pathways remain unclear. This review summarizes the cellular and molecular relationships between vascular biology and HL. Proteins associated with various functions in vascular biology, including cytokines (TNF-α, IL-1, IL-13, and IL-21), chemokines (CXCL10, CXCL12, and CCL21), adhesion molecules (ELAM-1/VCAM-1), and growth factors (BDNF/NT-3, platelet-derived growth factor receptor-α), have been linked to tumor activity. Notable tumor activities include the induction of paracrine activation of NF-kB-dependent pathways, upregulation of adhesion molecule regulation, genome amplification, and effective loss of antigen presentation mediated by MHC-II. Preclinical study models, primarily those using cell culture, have been optimized for HL. Animal models, particularly mice, are also used as alternatives to complex biological systems, with studies primarily focusing on the physiopathogenic evaluation of the disease. These biomolecules warrant further study because they may shed light on obscure pathways and serve as targets for prevention and/or treatment interventions.
Collapse
Affiliation(s)
- Wellington Francisco Rodrigues
- Postgraduate Course in Tropical Medicine and Infectious Diseases, Federal University of Triangulo Mineiro, UFTM, Uberaba 38025-440, MG, Brazil; (C.B.M.); (C.J.F.O.)
- University Center of Mineiros, Unifimes, Mineiros 75833-130, GO, Brazil; (M.C.M.d.A.); (J.M.N.)
| | - Camila Botelho Miguel
- Postgraduate Course in Tropical Medicine and Infectious Diseases, Federal University of Triangulo Mineiro, UFTM, Uberaba 38025-440, MG, Brazil; (C.B.M.); (C.J.F.O.)
- University Center of Mineiros, Unifimes, Mineiros 75833-130, GO, Brazil; (M.C.M.d.A.); (J.M.N.)
| | | | - Jamil Miguel Neto
- University Center of Mineiros, Unifimes, Mineiros 75833-130, GO, Brazil; (M.C.M.d.A.); (J.M.N.)
| | - Carlo José Freire Oliveira
- Postgraduate Course in Tropical Medicine and Infectious Diseases, Federal University of Triangulo Mineiro, UFTM, Uberaba 38025-440, MG, Brazil; (C.B.M.); (C.J.F.O.)
| |
Collapse
|
37
|
Ibáñez-Navarro M, Fernández A, Escudero A, Esteso G, Campos-Silva C, Navarro-Aguadero MÁ, Leivas A, Caracuel BR, Rodríguez-Antolín C, Ortiz A, Navarro-Zapata A, Mestre-Durán C, Izquierdo M, Balaguer-Pérez M, Ferreras C, Martínez-López J, Valés-Gómez M, Pérez-Martínez A, Fernández L. NKG2D-CAR memory T cells target pediatric T-cell acute lymphoblastic leukemia in vitro and in vivo but fail to eliminate leukemia initiating cells. Front Immunol 2023; 14:1187665. [PMID: 37928520 PMCID: PMC10622787 DOI: 10.3389/fimmu.2023.1187665] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Refractory/relapsed pediatric acute leukemia are still clinically challenging and new therapeutic strategies are needed. Interactions between Natural Killer Group 2D (NKG2D) receptor, expressed in cytotoxic immune cells, and its ligands (NKG2DL), which are upregulated in leukemic blasts, are important for anti-leukemia immunosurveillance. Nevertheless, leukemia cells may develop immunoescape strategies as NKG2DL shedding and/or downregulation. Methods In this report, we analyzed the anti-leukemia activity of NKG2D chimeric antigen receptor (CAR) redirected memory (CD45RA-) T cells in vitro and in a murine model of T-cell acute lymphoblastic leukemia (T-ALL). We also explored in vitro how soluble NKG2DL (sNKG2DL) affected NKG2D-CAR T cells' cytotoxicity and the impact of NKG2D-CAR T cells on Jurkat cells gene expression and in vivo functionality. Results In vitro, we found NKG2D-CAR T cells targeted leukemia cells and showed resistance to the immunosuppressive effects exerted by sNKG2DL. In vivo, NKG2D-CAR T cells controlled T cell leukemia burden and increased survival of the treated mice but failed to cure the animals. After CAR T cell treatment, Jurkat cells upregulated genes related to proliferation, survival and stemness, and in vivo, they exhibited functional properties of leukemia initiating cells. Discussion The data here presented suggest, that, in combination with other therapeutic approaches, NKG2D-CAR T cells could be a novel treatment for pediatric T-ALL.
Collapse
Affiliation(s)
- Marta Ibáñez-Navarro
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Adrián Fernández
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Adela Escudero
- Pediatric Oncology Department, Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, Madrid, Spain
| | - Gloria Esteso
- Tumor Immune Activation and Evasion Lab. Immunology and Oncology Department, National Biotechnology Center (CNB), Madrid, Spain
| | - Carmen Campos-Silva
- Tumor Immune Activation and Evasion Lab. Immunology and Oncology Department, National Biotechnology Center (CNB), Madrid, Spain
| | - Miguel Ángel Navarro-Aguadero
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alejandra Leivas
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Beatriz Ruz Caracuel
- Pediatric Oncology Department, Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, Madrid, Spain
| | - Carlos Rodríguez-Antolín
- Biomarkers and Experimental Therapeutics in Cancer, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
- Cancer Epigenetics Laboratory, Genetic Unit, Hospital Universitario La Paz, Madrid, Spain
| | - Alejandra Ortiz
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alfonso Navarro-Zapata
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Carmen Mestre-Durán
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Manuel Izquierdo
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - María Balaguer-Pérez
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristina Ferreras
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Joaquín Martínez-López
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Hematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Mar Valés-Gómez
- Tumor Immune Activation and Evasion Lab. Immunology and Oncology Department, National Biotechnology Center (CNB), Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- Pediatric Hemato-Oncology, Hospital Universitario La Paz, Madrid, Spain
- Pediatric Department, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lucía Fernández
- Hematological Malignancies-H12O Lab. Clinical Research Department, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
38
|
Tam OCH, Ho RSL, Chan S, Li KKW, Lam TL, Cheung ETY, Cheung OY, Ho WWS, Cheng KKF, Shing MMK, Ku DTL, Chung BHY, Yang W, Chan GCF, Ng HK, Liu APY. Genome-Wide DNA Methylation Profiling as Frontline Diagnostics for Central Nervous System Embryonal Tumors in Hong Kong. Cancers (Basel) 2023; 15:4880. [PMID: 37835574 PMCID: PMC10571663 DOI: 10.3390/cancers15194880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
This paper examines the link between CNS tumor biology and heterogeneity and the use of genome-wide DNA methylation profiling as a clinical diagnostic platform. CNS tumors are the most common solid tumors in children, and their prognosis remains poor. This study retrospectively analyzed pediatric patients with CNS embryonal tumors in Hong Kong between 1999 and 2017, using data from the territory-wide registry and available formalin-fixed paraffin-embedded tumor tissue. After processing archival tumor tissue via DNA extraction, quantification, and methylation profiling, the data were analyzed by using the web-based DKFZ classifier (Molecular Neuropathology (MNP) 2.0 v11b4) and t-SNE analysis. Methylation profiles were deemed informative in 85 samples. Epigenetic data allowed molecular subgrouping and confirmed diagnosis in 65 samples, verified histologic diagnosis in 8, and suggested an alternative diagnosis in 12. This study demonstrates the potential of DNA methylation profiling in characterizing pediatric CNS embryonal tumors in a large cohort from Hong Kong, which should enable regional and international collaboration in future pediatric neuro-oncology research.
Collapse
Affiliation(s)
- Otto C. H. Tam
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (O.C.H.T.)
| | - Ronnie S. L. Ho
- Department of Pathology, Gleneagles Hospital, Wong Chuk Hang, Hong Kong
| | - Shing Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (O.C.H.T.)
| | - Kay K. W. Li
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Tit-Leung Lam
- Department of Pathology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | | | - Oi-Yee Cheung
- Department of Pathology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Wilson W. S. Ho
- Department of Neurosurgery, Queen Mary Hospital, Pok Fu Lam, Hong Kong
- Department of Neurosurgery, Hong Kong Children’s Hospital, Kowloon, Hong Kong
| | - Kevin K. F. Cheng
- Department of Neurosurgery, Queen Mary Hospital, Pok Fu Lam, Hong Kong
- Department of Neurosurgery, Hong Kong Children’s Hospital, Kowloon, Hong Kong
| | - Matthew M. K. Shing
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Kowloon, Hong Kong
| | - Dennis T. L. Ku
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Kowloon, Hong Kong
| | - Brian H. Y. Chung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (O.C.H.T.)
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (O.C.H.T.)
| | - Godfrey C. F. Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (O.C.H.T.)
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Kowloon, Hong Kong
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Anthony P. Y. Liu
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong; (O.C.H.T.)
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Kowloon, Hong Kong
| |
Collapse
|
39
|
Tosi U, Souweidane M. Fifty years of DIPG: looking at the future with hope. Childs Nerv Syst 2023; 39:2675-2686. [PMID: 37382660 DOI: 10.1007/s00381-023-06037-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a primary brainstem tumor of childhood that carries a dismal prognosis, with median survival of less than 1 year. Because of the brain stem location and pattern of growth within the pons, Dr. Harvey Cushing, the father of modern neurosurgery, urged surgical abandonment. Such a dismal prognosis remained unchanged for decades, coupled with a lack of understanding of tumor biology and an unchanging therapeutic panorama. Beyond palliative external beam radiation therapy, no therapeutic approach has been widely accepted. In the last one to two decades, however, increased tissue availability, an improving understanding of biology, genetics, and epigenetics have led to the development of novel therapeutic targets. In parallel with this biological revolution, new methods intended to enhance drug delivery into the brain stem are contributing to a surge of exciting experimental therapeutic strategies.
Collapse
Affiliation(s)
- Umberto Tosi
- Department of Neurosurgery, Weill Cornell Medicine, 525 E 68th St Box 99, New York, NY, 10021, USA
| | - Mark Souweidane
- Department of Neurosurgery, Weill Cornell Medicine, 525 E 68th St Box 99, New York, NY, 10021, USA.
| |
Collapse
|
40
|
Hsieh PL, Tsai KL, Chou WC, Wu CH, Jou IM, Tu YK, Ma CH. Cisplatin triggers oxidative stress, apoptosis and pro-inflammatory responses by inhibiting the SIRT1-mediated Nrf2 pathway in chondrocytes. ENVIRONMENTAL TOXICOLOGY 2023; 38:2476-2486. [PMID: 37497868 DOI: 10.1002/tox.23885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 07/28/2023]
Abstract
Although the height of the proliferating layer that was suppressed in the growth plate has been recognized as an adverse effect of cisplatin in pediatric cancer survivors, the detailed pathological mechanism has not been elucidated. Sirtuin-1 (SIRT1) has been reported as an essential modulator of cartilage homeostasis, but its role in cisplatin-induced damage of chondrocytes remains unclear. In this study, we examined how cisplatin affected the expression of SIRT1 and cell viability. Next, we showed downregulation of SIRT1 after cisplatin treatment resulted in suppression of Peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α), leading to inhibition of Nrf2 nuclear translocation and subsequently decreased Heme oxygenase-1(HO-1) and NAD(P)H Quinone Dehydrogenase 1(NQO-1) expression. Blockage of the SIRT1/ PGC-1α axis not only increased oxidative stress with lower antioxidant SOD and GSH, but also contributed to mitochondrial dysfunction evidenced by the collapse of membrane potential and repression of mitochondrial DNA copy number and ATP. We also found that Cisplatin up-regulated the p38 phosphorylation, pro-inflammatory events and matrix metalloproteinases (MMPs) in chondrocytes through the SIRT1-modulated antioxidant manner. Collectively, our findings suggest that preservation of SIRT1 in chondrocytes may be a potential target to ameliorate growth plate dysfunction for cisplatin-receiving pediatric cancer survivors.
Collapse
Affiliation(s)
- Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Ching Chou
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan
| | - Chin-Hsien Wu
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan
| | - I-Ming Jou
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan
| | - Ching-Hou Ma
- Department of Orthopedics, E-Da Hospital, I-Shou University, Kaohsiung City, Taiwan
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung City, Taiwan
| |
Collapse
|
41
|
Ha L, Wakefield CE, Diaz C, Mizrahi D, Signorelli C, Yacef K, Simar D. Patterns of physical activity and sedentary behavior in child and adolescent cancer survivors assessed using wrist accelerometry: A cluster analysis approach. Health Informatics J 2023; 29:14604582231212525. [PMID: 37903362 DOI: 10.1177/14604582231212525] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Physical activity levels among childhood cancer survivors are typically quantified as a total amount using time spent in various intensities. Yet, most analyses do not consider the transitory nature of children's behaviors and a more detailed approach could provide complimentary information. We aimed to explore various behavior profiles of survivors' daily and hourly physical activity patterns. We measured 8-18-year-old survivors' activity levels over 7 days using wrist accelerometry and cluster analysis. Of the 37 participant datasets, survivors engaged in mean (SD) 36.3 (19.0) min/day of MVPA and 4.1 (1.9) hrs/day of sedentary activity. The cluster analysis revealed five daily movement patterns: 'most active' (prevalence 11%), 'active' (22%), 'moderately active + moderately sedentary' (35%), 'moderately active + high sedentary' (5%) and 'least active' (27%). Younger survivors and those with less time since treatment completion were more likely to be in the active clusters. Hourly behaviors were characterized by short bursts of MVPA and moderate bouts of sedentary activity. Our approach provides an insightful analysis into the nature and timing of childhood cancer survivors' movement behaviours. Our findings may assist in the development of targeted interventions to improve physical activity levels.
Collapse
Affiliation(s)
- Lauren Ha
- School of Health Sciences, Faculty of Medicine & Health, UNSW, Sydney, Australia
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Claire E Wakefield
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
- School of Clinical Medicine, Discipline of Paediatrics & Child Health, Faculty of Medicine & Health, UNSW, Sydney, Australia
| | - Claudio Diaz
- School of Computer Science, The University of Sydney, Australia
| | - David Mizrahi
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council, NSW, Australia
- Prince of Wales Clinical School, Faculty of Medicine & Health, UNSW Sydney, Australia
| | - Christina Signorelli
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
- School of Clinical Medicine, Discipline of Paediatrics & Child Health, Faculty of Medicine & Health, UNSW, Sydney, Australia
| | - Kalina Yacef
- School of Computer Science, The University of Sydney, Australia
| | - David Simar
- School of Health Sciences, Faculty of Medicine & Health, UNSW, Sydney, Australia
| |
Collapse
|
42
|
Madi D, Abi Abdallah Doumit M, Hallal M, Moubarak MM. Outlooks on using a mobile health intervention for supportive pain management for children and adolescents with cancer: a qualitative study. BMC Nurs 2023; 22:301. [PMID: 37667338 PMCID: PMC10476416 DOI: 10.1186/s12912-023-01461-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Considerable improvements in the prognosis of pediatric cancer patients have been achieved over recent decades due to advances in treatment. Nevertheless, as the most common and distressing health issue for pediatrics with cancer, cancer-related pain is still a significant hurdle that impedes patients' journey to recovery, compromises their quality of life, and delays the positive outcome and effectiveness of their treatments. PURPOSE Taking into consideration that acceptability studies are imperative for the design, evaluation, and implementation of healthcare interventions, this study aims to explore pediatric oncology patients' readiness to use a mobile health application that emphasizes social assistance and peer support in addition to conventional pain management methods. DESIGN AND METHODS This study followed the Qualitative description approach. Twelve participants were chosen based on purposive sampling and maximum variation sampling. Interviews were analyzed using the conventional content analysis. RESULTS Analysis of the interviews revealed four major categories: (A) The need for connectedness; (B) An innovative way to connect yet fearful; (C) A 3D approach; (D) Fears of the unfamiliar. CONCLUSIONS This study is the first in Lebanon and the region to undertake an initiative towards introducing technology for pain assessment and management of children with cancer through a dedicated digital platform. The study results attested to the acceptability and potential utilization of this platform by children with cancer. PRACTICE IMPLICATIONS Nurses need to be trained to play an essential role in teaching children with cancer about the significance of social support and assisting them to establish their social support network. Children with cancer are encouraged to voice out their need for help. Our proposed application can create an enabling environment to harness the power of social support and provide children with cancer the opportunity to connect on a deeper level in a supportive and pity-free space.
Collapse
Affiliation(s)
- Dina Madi
- Hariri School of Nursing, American University of Beirut, Beirut, Lebanon
| | | | - Mohammad Hallal
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maya M Moubarak
- Hariri School of Nursing, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
43
|
Kourti M, Aivaliotis M, Hatzipantelis E. Proteomics in Childhood Acute Lymphoblastic Leukemia: Challenges and Opportunities. Diagnostics (Basel) 2023; 13:2748. [PMID: 37685286 PMCID: PMC10487225 DOI: 10.3390/diagnostics13172748] [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: 06/30/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children and one of the success stories in cancer therapeutics. Risk-directed therapy based on clinical, biologic and genetic features has played a significant role in this accomplishment. Despite the observed improvement in survival rates, leukemia remains one of the leading causes of cancer-related deaths. Implementation of next-generation genomic and transcriptomic sequencing tools has illustrated the genomic landscape of ALL. However, the underlying dynamic changes at protein level still remain a challenge. Proteomics is a cutting-edge technology aimed at deciphering the mechanisms, pathways, and the degree to which the proteome impacts leukemia subtypes. Advances in mass spectrometry enable high-throughput collection of global proteomic profiles, representing an opportunity to unveil new biological markers and druggable targets. The purpose of this narrative review article is to provide a comprehensive overview of studies that have utilized applications of proteomics in an attempt to gain insight into the pathogenesis and identification of biomarkers in childhood ALL.
Collapse
Affiliation(s)
- Maria Kourti
- Third Department of Pediatrics, School of Medicine, Aristotle University and Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michalis Aivaliotis
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Emmanouel Hatzipantelis
- Children & Adolescent Hematology-Oncology Unit, Second Department of Pediatrics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
44
|
Banwait DK, Arora PR, Mahajan A, Dinand V, Jain S, Kalra M, Chandra J, Arora RS. Barriers to Accessing Fertility Preservation in Adolescents with Hodgkin Lymphoma in India. Pediatr Hematol Oncol 2023; 41:163-168. [PMID: 37264813 DOI: 10.1080/08880018.2023.2218444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 05/21/2023] [Indexed: 06/03/2023]
Affiliation(s)
| | | | - Amita Mahajan
- Department of Pediatric Oncology, Indraprastha Apollo Hospital, New Delhi, India
| | - Veronique Dinand
- Palliative and Supportive Care Unit, Bai Jerbai Wadia Hospital, Mumbai, India
| | - Sandeep Jain
- Department of Pediatric Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| | - Manas Kalra
- Pediatric Hematology, Oncology BMT Unit, Sir Ganga Ram Hospital, New Delhi, India
| | - Jagdish Chandra
- Department of Pediatrics, PGIMSR and ESIC Model Hospital, Basaidarapur, New Delhi, India
| | - Ramandeep Singh Arora
- Max Institute of Cancer Care, Max Super Speciality Hospital, Saket, New Delhi, India
| |
Collapse
|
45
|
Mirutse MK, Palm MT, Tolla MT, Memirie ST, Kefyalew ES, Hailu D, Norheim OF. Cost of childhood cancer treatment in Ethiopia. PLoS One 2023; 18:e0286461. [PMID: 37267276 DOI: 10.1371/journal.pone.0286461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 05/09/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Despite the recent interest in expanding pediatric oncology units in Ethiopia, reflected in the National Childhood and Adolescent Cancer Control Plan (NCACCP), little is known about the cost of running a pediatric oncology unit and treating childhood cancers. METHODS We collected historical cost data and quantity of services provided for the pediatric oncology unit and all other departments in Tikur Anbessa Specialized Hospital (TASH) from 8 July 2018 to 7 July 2019, using a provider perspective and mixed (top-down and bottom-up) costing approaches. Direct costs (human resources, drugs, supplies, medical equipment) of the pediatric oncology unit, costs at other relevant clinical departments, and overhead cost share are summed up to estimate the total annual cost of running the unit. Further, unit costs were estimated at specific childhood cancer levels. RESULTS The estimated annual total cost of running a pediatric oncology unit was USD 776,060 (equivalent to USD 577 per treated child). The cost of running a pediatric oncology unit per treated child ranged from USD 469 to USD 1,085, on the scenario-based sensitivity analysis. Drugs and supplies, and human resources accounted for 33% and 27% of the total cost, respectively. Outpatient department and inpatient department shared 37% and 63% of the cost, respectively. For the pediatric oncology unit, the cost per OPD visit, cost per bed day, and cost per episode of hospital admission were USD 36.9, 39.9, and 373.3, respectively. The annual cost per treated child ranged from USD 322 to USD 1,313 for the specific childhood cancers. CONCLUSION Running a pediatric oncology unit in Ethiopia is likely to be affordable. Further analysis of cost effectiveness, equity, and financial risk protection impacts of investing in childhood cancer programs could better inform the prioritization of childhood cancer control interventions in the Ethiopia Essential Health Service Package.
Collapse
Affiliation(s)
- Mizan Kiros Mirutse
- Department of Global Public Health and Primary Care, Bergen Centre for Ethics and Priority Setting (BCEPS), University of Bergen, Bergen, Norway
- Ministry of Health Ethiopia, Addis Ababa, Ethiopia
| | | | - Mieraf Taddesse Tolla
- Department of Global Public Health and Primary Care, Bergen Centre for Ethics and Priority Setting (BCEPS), University of Bergen, Bergen, Norway
| | - Solomon Tessema Memirie
- Department of Global Public Health and Primary Care, Bergen Centre for Ethics and Priority Setting (BCEPS), University of Bergen, Bergen, Norway
- Addis Center for Ethics and Priority Setting, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Daniel Hailu
- Department of Pediatrics and Child Health, Pediatric Hematology/Oncology Unit, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Ole F Norheim
- Department of Global Public Health and Primary Care, Bergen Centre for Ethics and Priority Setting (BCEPS), University of Bergen, Bergen, Norway
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America
| |
Collapse
|
46
|
Jain D, Somasundaram DB, Aravindan S, Yu Z, Baker A, Esmaeili A, Aravindan N. Prognostic significance of NT5E/CD73 in neuroblastoma and its function in CSC stemness maintenance. Cell Biol Toxicol 2023; 39:967-989. [PMID: 34773529 DOI: 10.1007/s10565-021-09658-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/10/2021] [Indexed: 12/22/2022]
Abstract
Cluster of differentiation 73 (CD73), a cell surface enzyme that catalyzes adenosine monophosphate (AMP) breakdown to adenosine, is differentially expressed in cancers and has prognostic significance. We investigated its expression profile in neuroblastoma (NB), its association with NB clinical outcomes, and its influence in the regulation of cancer stem cells' (CSCs) stemness maintenance. RNA-Seq data mining (22 independent study cohorts, total n = 3836) indicated that high CD73 can predict good NB prognosis. CD73 expression (immunohistochemistry) gauged in an NB patient cohort (n = 87) showed a positive correlation with longer overall survival (OS, P = 0.0239) and relapse-free survival (RFS, P = 0.0242). Similarly, high CD73 correlated with longer OS and RFS in advanced disease stages, MYCN non-amplified (MYCN-na), and Stage-4-MYCN-na subsets. Despite no definite association in children < 2 years old (2Y), high CD73 correlated with longer OS (P = 0.0294) and RFS (P = 0.0315) in children > 2Y. Consistently, high CD73 was associated with better OS in MYCN-na, high-risk, and stage-4 subsets of children > 2Y. Multivariate analysis identified CD73 as an independent (P = 0.001) prognostic factor for NB. Silencing CD73 in patient-derived (stage 4, progressive disease) CHLA-171 and CHLA-172 cells revealed cell-line-independent activation of 58 CSC stemness maintenance molecules (QPCR profiling). Overexpressing CD73 in CHLA-20 and CHLA-90 cells with low CD73 and silencing in CHLA-171 and CHLA-172 cells with high CD73 showed that CD73 regulates epithelial to mesenchymal transition (E-Cadherin, N-Cadherin, Vimentin), stemness maintenance (Sox2, Nanog, Oct3/4), self-renewal capacity (Notch), and differentiation inhibition (leukemia inhibitory factor, LIF) proteins (confocal-immunofluorescence). These results demonstrate that high CD73 can predict good prognosis in NB, and further suggest that CD73 regulates stemness maintenance in cells that defy clinical therapy.
Collapse
Affiliation(s)
- Drishti Jain
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dinesh Babu Somasundaram
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Zhongxin Yu
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ashley Baker
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Azadeh Esmaeili
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Natarajan Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, Oklahoma City, OK, USA.
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
47
|
Zhang Q, Huang MJ, Wang HY, Wu Y, Chen YZ. A novel prognostic nomogram for adult acute lymphoblastic leukemia: a comprehensive analysis of 321 patients. Ann Hematol 2023:10.1007/s00277-023-05267-6. [PMID: 37173535 DOI: 10.1007/s00277-023-05267-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/06/2023] [Indexed: 05/15/2023]
Abstract
The cure rate of acute lymphoblastic leukemia (ALL) in adolescents and adults remains poor. This study aimed to establish a prognostic model for ≥14-year-old patients with ALL to guide treatment decisions. We retrospectively analyzed the data of 321 ALL patients between January 2017 and June 2020. Patients were randomly (2:1 ratio) divided into either the training or validation set. A nomogram was used to construct a prognostic model. Multivariate Cox analysis of the training set showed that age > 50 years, white blood cell count > 28.52×109/L, and MLL rearrangement were independent risk factors for overall survival (OS), while platelet count >37×109/L was an independent protective factor. The nomogram was established according to these independent prognostic factors in the training set, where patients were grouped into two categories: low-risk (≤13.15) and high-risk (>13.15). The survival analysis, for either total patients or sub-group patients, showed that both OS and progression-free survival (PFS) of low-risk patients was significantly better than that of high-risk patients. Moreover, treatment analysis showed that both OS and progression-free survival (PFS) of ALL with stem cell transplantation (SCT) were significantly better than that of ALL without SCT. Further stratified analysis showed that in low-risk patients, the OS and PFS of patients with SCT were significantly better than those of patients without SCT. In contrast, in high-risk patients, compared with non-SCT patients, receiving SCT can only significantly prolong the PFS, but it does not benefit the OS. We established a simple and effective prognostic model for ≥ 14-year-old patients with ALL that can provide accurate risk stratification and determine the clinical strategy.
Collapse
Affiliation(s)
- Qian Zhang
- Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mei-Juan Huang
- Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Han-Yu Wang
- Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yong Wu
- Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Yuan-Zhong Chen
- Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
48
|
Berko ER, Witek GM, Matkar S, Petrova ZO, Wu MA, Smith CM, Daniels A, Kalna J, Kennedy A, Gostuski I, Casey C, Krytska K, Gerelus M, Pavlick D, Ghazarian S, Park JR, Marachelian A, Maris JM, Goldsmith KC, Radhakrishnan R, Lemmon MA, Mossé YP. Circulating tumor DNA reveals mechanisms of lorlatinib resistance in patients with relapsed/refractory ALK-driven neuroblastoma. Nat Commun 2023; 14:2601. [PMID: 37147298 PMCID: PMC10163008 DOI: 10.1038/s41467-023-38195-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/20/2023] [Indexed: 05/07/2023] Open
Abstract
Activating point mutations in Anaplastic Lymphoma Kinase (ALK) have positioned ALK as the only mutated oncogene tractable for targeted therapy in neuroblastoma. Cells with these mutations respond to lorlatinib in pre-clinical studies, providing the rationale for a first-in-child Phase 1 trial (NCT03107988) in patients with ALK-driven neuroblastoma. To track evolutionary dynamics and heterogeneity of tumors, and to detect early emergence of lorlatinib resistance, we collected serial circulating tumor DNA samples from patients enrolled on this trial. Here we report the discovery of off-target resistance mutations in 11 patients (27%), predominantly in the RAS-MAPK pathway. We also identify newly acquired secondary compound ALK mutations in 6 (15%) patients, all acquired at disease progression. Functional cellular and biochemical assays and computational studies elucidate lorlatinib resistance mechanisms. Our results establish the clinical utility of serial circulating tumor DNA sampling to track response and progression and to discover acquired resistance mechanisms that can be leveraged to develop therapeutic strategies to overcome lorlatinib resistance.
Collapse
Affiliation(s)
- Esther R Berko
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Pediatric Hematology and Oncology, Schneider Children's Medical Center, Petach Tikva, Israel, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gabriela M Witek
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Smita Matkar
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zaritza O Petrova
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Megan A Wu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Courtney M Smith
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Alex Daniels
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joshua Kalna
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Annie Kennedy
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ivan Gostuski
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Colleen Casey
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kateryna Krytska
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark Gerelus
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Susan Ghazarian
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julie R Park
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Araz Marachelian
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly C Goldsmith
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
- Seattle Children's Hospital, Seattle, WA, USA
| | - Ravi Radhakrishnan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark A Lemmon
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA.
| | - Yaël P Mossé
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Petricca K, Kambugu J, Githang'a J, Macharia WM, Njuguna F, McLigeyo A, Nyangasi M, Orem J, Kanyamuhunga A, Laiti R, Katabalo D, Schroeder K, Rogo K, Maguire B, Wambui L, Nkurunziza JN, Wong B, Neposlan J, Kilawe L, Gupta S, Denburg AE. Access to essential cancer medicines for children: a comparative mixed-methods analysis of availability, price, and health-system determinants in east Africa. Lancet Oncol 2023; 24:563-576. [PMID: 37023781 DOI: 10.1016/s1470-2045(23)00072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Access to essential childhood cancer medicines is a core determinant of childhood cancer outcomes. Available evidence, although scarce, suggests that access to these medicines is highly variable across countries, particularly in low-income and middle-income countries, where the burden of childhood cancer is greatest. To support evidence-informed national and regional policies for improved childhood cancer outcomes, we aimed to analyse access to essential childhood cancer medicines in four east African countries-Kenya, Rwanda, Tanzania, and Uganda-by determining the availability and price of these medicines and the health system determinants of access. METHODS In this comparative analysis, we used prospective mixed-method analyses to track and analyse the availability and price of essential childhood cancer medicines, investigate contextual determinants of access to childhood cancer medicines within and across included countries, and assess the potential effects of medicine stockouts on treatment. Eight tertiary care hospitals were included, seven were public sites (Kenyatta National Hospital [KNH; Nairobi, Kenya], Jaramogi Oginga Odinga Referral and Teaching Hospital [JOORTH; Kisumu, Kenya], Moi University Teaching and Referral Hospital [MTRH; Eldoret, Kenya], Bugando Medical Centre [BMC; Mwanza, Tanzania], Muhimbili National Hospital [MNH; Dar es Salaam, Tanzania], Butaro Cancer Centre of Excellence [BCCE; Butaro Sector, Rwanda], and Uganda Cancer Institute [UCI; Kampala, Uganda]) and one was a private site (Aga Khan University Hospital [AKU; Nairobi, Kenya]). We catalogued prices and stockouts for 37 essential drugs from each of the eight study siteson the basis of 52 weeks of prospective data that was collected across sites from May 1, 2020, to Jan 31, 2022. We analysed determinants of medicine access using thematic analysis of academic literature, policy documents, and semi-structured interviews from a purposive sample of health system stakeholders. FINDINGS Recurrent stockouts of a wide range of cytotoxic and supportive care medicines were observed across sites, with highest mean unavailability in Kenya (JOORTH; 48·5%), Rwanda (BCCE; 39·0%), and Tanzania (BMC; 32·2%). Drugs that had frequent stockouts across at least four sites included methotrexate, bleomycin, etoposide, ifosfamide, oral morphine, and allopurinol. Average median price ratio of medicines at each site was within WHO's internationally accepted threshold for efficient procurement (median price ratio ≤1·5). The effect of stockouts on treatment was noted across most sites, with the greatest potential for treatment interruptions in patients with Hodgkin lymphoma, retinoblastoma, and acute lymphocytic leukaemia. Policy prioritisation of childhood cancers, health financing and coverage, medicine procurement and supply chain management, and health system infrastructure emerged as four prominent determinants of access when the stratified purposive sample of key informants (n=64) across all four countries (Kenya n=19, Rwanda n=15, Tanzania n=13, and Uganda n=17) was interviewed. INTERPRETATION Access to childhood cancer medicines across east Africa is marked by gaps in availability that have implications for effective treatment delivery for a range of childhood cancers. Our findings provide detailed evidence of barriers to access to childhood cancer medicine at multiple points in the pharmaceutical value chain. These data could inform national and regional policy makers to optimise cancer medicine availability and affordability as part of efforts to improve childhood cancer outcomes specific regions and internationally. FUNDING American Childhood Cancer Organization, Childhood Cancer International, and the Friends of Cancer Patients Ameera Fund.
Collapse
Affiliation(s)
- Kadia Petricca
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Joyce Kambugu
- Department of Paediatric Oncology, Uganda Cancer Institute, Kampala, Uganda
| | - Jessie Githang'a
- Department of Human Pathology, University of Nairobi, Nairobi, Kenya
| | - William M Macharia
- Department of Pediatrics and Child Health, Aga Khan University, Nairobi, Kenya
| | - Festus Njuguna
- Department of Child Health and Paediatrics, Moi University, Eldoret, Kenya
| | - Angela McLigeyo
- Jaramogi Oginga Odinga Teaching and Referral Hospital, Oncology, Kisumu, Kenya
| | - Mary Nyangasi
- National Cancer Control Program, Ministry of Health, Nairobi, Kenya
| | - Jackson Orem
- Department of Paediatric Oncology, Uganda Cancer Institute, Kampala, Uganda
| | - Aimable Kanyamuhunga
- College of Medicine and Health Sciences University of Rwanda, Child Health, Kigali, Rwanda
| | - Rehema Laiti
- Muhimbili National Hospital, Oncology, Dar Es Salaam, Tanzania
| | | | | | - Khama Rogo
- African Institute for Health Transformation, Luanda, Kenya
| | - Bryan Maguire
- Biostatistics, Design and Analysis, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lucy Wambui
- Institute of Anthropology and Gender Studies, University of Nairobi, Nairobi, Kenya
| | - Jean N Nkurunziza
- College of Medicine and Health Sciences University of Rwanda, Child Health, Kigali, Rwanda
| | - Bryan Wong
- School of Medicine, Queen's University, Kingston, ON, Canada
| | - Joshua Neposlan
- Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Lilian Kilawe
- Department of Public Health, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Sumit Gupta
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Avram E Denburg
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
50
|
Srinivasan S, Ramanathan S, Prasad M. Wilms Tumor in India: A Systematic Review. South Asian J Cancer 2023; 12:206-212. [PMID: 37969674 PMCID: PMC10635777 DOI: 10.1055/s-0042-1758567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Shyam SrinivasanBackground Cure rates of childhood malignancies are inferior in India compared with upper-middle-income countries. There is paucity of quality data addressing outcome of childhood Wilms tumor (WT) from India. This systematic review was conducted to assess the disease trends, treatment strategies, and outcome indicators in WT across India. Materials and Methods We conducted a systematic search of MEDLINE, Google Scholar, and SCOPUS database, and additionally screened International Society of Pediatric Oncology conference abstracts. Data concerning WT or nephroblastoma published from India were extracted. Results A total of 17 studies containing 1,170 patients were included in this review. Ninety-four percent of the studies were published after the year 2010. Advanced stage (III and IV) disease was seen in 46% of included patients. In seven studies, patients underwent a pretreatment biopsy before commencement of therapy. A hybrid approach consisting of "surgery first" in a selected subset and "neo-adjuvant chemotherapy" in all others was the most common treatment strategy adopted in half of the studies. The overall survival ranged between 48 and 89%. Key prognostic factors influencing survival across studies included increased tumor volume, metastatic disease, and unfavorable histology. Nonrelapse mortality (2.7-8.5%) was noted to be high. Conclusion Substantial proportion of children with WT from India present with advanced stages of the disease. Despite several limitations, the current systematic review showed a modest survival among Indian children with WT. Adopting strategies through collaboration to ensure early access to expert care along with involvement of social support team to improve compliance may further improve survival of WT in India.
Collapse
Affiliation(s)
- Shyam Srinivasan
- Department of Pediatric Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Subramaniam Ramanathan
- Department of Pediatric Oncology and BMT, Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, United Kingdom
| | - Maya Prasad
- Department of Pediatric Oncology, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, Maharashtra, India
| |
Collapse
|