1
|
Liu Z, Yuan Y, Wang N, Yu P, Teng Y. Drug combinations of camptothecin derivatives promote the antitumor properties. Eur J Med Chem 2024; 279:116872. [PMID: 39298971 DOI: 10.1016/j.ejmech.2024.116872] [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: 08/24/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Camptothecin (CPT) derivatives are widely used as small molecule chemotherapeutic agents and have demonstrated efficacy in the treatment of diverse solid tumors. A variety of derivatives have been developed to resolve the drawbacks of poor water solubility, high toxicity and rapid hydrolysis in vivo. However, the obstacles, such as acquired resistance and toxicity, still exist. The utilization of rational drug combinations has the potential to enhance the efficacy and mitigate the toxicity of CPT derivatives. This paper provides an overview of CPT derivatives in combination with other drugs, with a particular focus on cell cycle inhibitors, DNA synthesis inhibitors, anti-metastatic drugs and immunotherapy agents. Concurrently, the mechanisms of antitumor activity of combinations of different classes of drugs and CPT derivatives are elucidated. While the various combination strategies have yielded more favorable therapeutic outcomes, the efficacy and toxicity of the drug combinations are influenced by the inherent properties of the drugs involved. Moreover, a summary of the drug conjugates of CPT derivatives was provided, accompanied by an analysis of the structural activity relationship (SAR). This paves the way for the subsequent developments in drug combinations and delivery modes.
Collapse
Affiliation(s)
- Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| | - Yajie Yuan
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Ning Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
2
|
Rados M, Landegger A, Schmutzler L, Rabidou K, Taschner-Mandl S, Fetahu IS. Natural killer cells in neuroblastoma: immunological insights and therapeutic perspectives. Cancer Metastasis Rev 2024; 43:1401-1417. [PMID: 39294470 PMCID: PMC11554946 DOI: 10.1007/s10555-024-10212-8] [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: 04/15/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Natural killer (NK) cells have multifaceted roles within the complex tumor milieu. They are pivotal components of innate immunity and shape the dynamic landscape of tumor-immune cell interactions, and thus can be leveraged for use in therapeutic interventions. NK-based immunotherapies have had remarkable success in hematological malignancies, but these therapies are met with many challenges in solid tumors, including neuroblastoma (NB), a childhood tumor arising from the sympathetic nervous system. With a focus on NB, this review outlines the mechanisms employed by NK cells to recognize and eliminate malignant cells, delving into the dynamic relationship between ligand-receptor interactions, cytokines, and other molecules that facilitate the cross talk between NK and NB cells. We discuss the immunomodulatory functions of NK cells and the mechanisms that contribute to loss of this immunosurveillance in NB, with a focus on how this dynamic has been utilized in recent immunotherapy advancements for NB.
Collapse
Affiliation(s)
- Magdalena Rados
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | | | - Lukas Schmutzler
- Department of Otorhinolaryngology - Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Kimberlie Rabidou
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, USA
| | | | - Irfete S Fetahu
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Wu H, Zhang G, Liu Z, Liu W, Wang X, Zhao Y. Enhanced anti-tumor activity mediated by combination chimeric antigen receptor T cells targeting GD2 and GPC2 in high-risk neuroblastoma. Cytotherapy 2024; 26:1308-1319. [PMID: 38904586 DOI: 10.1016/j.jcyt.2024.05.023] [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: 01/26/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND AIMS Chimeric antigen receptor T (CAR-T) cells targeting single antigens show limited activity against solid tumors due to poor T cell persistence, low efficiency infiltration, and exhaustion together with heterogeneous tumor-associated antigen (TAA) expression. This is also true in high-risk neuroblastoma (HRNB), a lethal pediatric extracranial malignancy. To overcome these obstacles, a combinational strategy using GD2-specific and GPC2-specific CAR-T cells was developed to improve immunotherapeutic efficacy. METHODS We individually developed GD2-specific and GPC2-specific CARs containing a selective domain (sCAR) which was a peptide of 10 amino acids derived from human nuclear autoantigen La/SS-B. These constructs allowed us to generate two different HRNB antigen-specific CAR-T cells with enhanced biological activity through stimulating sCAR-engrafted T cells via a selective domain-specific monoclonal antibody (SmAb). Binding affinity and stimulation of GD2- and GPC2-specific sCARs by SmAb were measured, and transient and persistent anti-tumor cytotoxicity of GD2sCAR-T and GPC2sCAR-T cells were quantified in neuroblastoma cell lines expressing different TAA levels. The anti-tumor pharmaceutical effects and cellular mechanisms mediated by single or combinational sCAR-T cells were evaluated in vitro and in vivo. RESULTS GD2- and GPC2-specific sCARs had antigen-specific binding affinity similar to their parental counterparts and were recognized by SmAb. SmAb-mediated stimulation selectively activated sCAR-T proliferation and increased central memory T cells in the final products. SmAb-stimulated sCAR-T cells had enhanced transient cytolytic activity, and combination therapy extended long-term anti-tumor activity in vitro through TNF-α and IL-15 release. Stimulated sCAR-T cells overcame heterogeneous antigen expression in HRNB, and the multi-TAA-targeting strategy was especially efficacious in vivo, inducing apoptosis through the caspase-3/PARP pathway and inhibiting the release of several tumor-promoting cytokines. CONCLUSIONS These data suggest that combined targeting of multiple TAAs is a promising strategy to overcome heterogenous antigen expression in solid tumors and extend CAR-T cell persistence for HRNB immunotherapy.
Collapse
Affiliation(s)
- Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Guangji Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Weihua Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Xuan Wang
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
4
|
Tempora P, D'Amico S, Gragera P, Damiani V, Krol K, Scaldaferri V, Pandey K, Chung S, Lucarini V, Giorda E, Scarsella M, Volpe G, Pezzullo M, De Stefanis C, D'Oria V, De Angelis L, Giovannoni R, De Ioris MA, Melaiu O, Purcell AW, Locatelli F, Fruci D. Combining ERAP1 silencing and entinostat therapy to overcome resistance to cancer immunotherapy in neuroblastoma. J Exp Clin Cancer Res 2024; 43:292. [PMID: 39438988 PMCID: PMC11494811 DOI: 10.1186/s13046-024-03180-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: 04/24/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Checkpoint immunotherapy unleashes tumor control by T cells, but it is undermined in non-immunogenic tumors, e.g. with low MHC class I expression and low neoantigen burden, such as neuroblastoma (NB). Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an enzyme that trims peptides before loading on MHC class I molecules. Inhibition of ERAP1 results in the generation of new antigens able of inducing potent anti-tumor immune responses. Here, we identify a novel non-toxic combinatorial strategy based on genetic inhibition of ERAP1 and administration of the HDAC inhibitor (HDACi) entinostat that increase the immunogenicity of NB, making it responsive to PD-1 therapy. METHODS CRISPR/Cas9-mediated gene editing was used to knockout (KO) the ERAP1 gene in 9464D NB cells derived from spontaneous tumors of TH-MYCN transgenic mice. The expression of MHC class I and PD-L1 was evaluated by flow cytometry (FC). The immunopeptidome of these cells was studied by mass spectrometry. Cocultures of splenocytes derived from 9464D bearing mice and tumor cells allowed the assessment of the effect of ERAP1 inhibition on the secretion of inflammatory cytokines and activation and migration of immune cells towards ERAP1 KO cells by FC. Tumor cell killing was evaluated by Caspase 3/7 assay and flow cytometry analysis. The effect of ERAP1 inhibition on the immune content of tumors was analyzed by FC, immunohistochemistry and multiple immunofluorescence. RESULTS We found that inhibition of ERAP1 makes 9464D cells more susceptible to immune cell-mediated killing by increasing both the recall and activation of CD4+ and CD8+ T cells and NK cells. Treatment with entinostat induces the expression of MHC class I and PD-L1 molecules in 9464D both in vitro and in vivo. This results in pronounced changes in the immunopeptidome induced by ERAP1 inhibition, but also restrains the growth of ERAP1 KO tumors in vivo by remodelling the tumor-infiltrating T-cell compartment. Interestingly, the absence of ERAP1 in combination with entinostat and PD-1 blockade overcomes resistance to PD-1 immunotherapy and increases host survival. CONCLUSIONS These findings demonstrate that ERAP1 inhibition combined with HDACi entinostat treatment and PD-1 blockade remodels the immune landscape of a non-immunogenic tumor such as NB, making it responsive to checkpoint immunotherapy.
Collapse
Affiliation(s)
| | | | - Paula Gragera
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Kamila Krol
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Kirti Pandey
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Shanzou Chung
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | | | - Ezio Giorda
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | | | | | | | | | | | | | | - Ombretta Melaiu
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Franco Locatelli
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Doriana Fruci
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
5
|
He Y, Wang J, Xiao T. Targeting the ubiquitin-proteasome system: a novel therapeutic strategy for neuroblastoma. Front Oncol 2024; 14:1443256. [PMID: 39391247 PMCID: PMC11464458 DOI: 10.3389/fonc.2024.1443256] [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: 06/03/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Neuroblastoma (NB) stands as a common and formidable malignant tumor among children, characterized by marked tumor heterogeneity and resistance to conventional treatments. Central to the regulation of protein stability, localization, and function is the process of ubiquitination-a critical protein modification. The therapeutic potential of drugs that target deubiquitination, demonstrated in the treatment of refractory multiple myeloma, warrants investigation in the context of NB. This review endeavors to demystify the intricate biological implications of ubiquitination within NB pathology, synthesize the current landscape of preclinical studies focused on the inhibition of the ubiquitin-proteasome system in NB, and assess the viability of this strategy as an innovative therapeutic frontier.
Collapse
Affiliation(s)
- Yangshen He
- Department of General Medicine, Dongguan Hospital of Integrated Chinese and Western Medicine, Dongguan, China
| | - Jianing Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tiantian Xiao
- Department of General Medicine, Dongguan Hospital of Integrated Chinese and Western Medicine, Dongguan, China
| |
Collapse
|
6
|
Sainero-Alcolado L, Bexelius TS, Santopolo G, Yuan Y, Liaño-Pons J, Arsenian-Henriksson M. Defining Neuroblastoma: from origin to precision medicine. Neuro Oncol 2024:noae152. [PMID: 39101440 DOI: 10.1093/neuonc/noae152] [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: 03/09/2024] [Indexed: 08/06/2024] Open
Abstract
Neuroblastoma (NB), an heterogenous pediatric tumor of the sympathetic nervous system, is the most common and deadly extracranial solid malignancy diagnosed in infants. Numerous efforts have been invested in understanding its origin and in development of novel curative targeted therapies. Here, we summarize the recent advances in the identification of the cell of origin and the genetic alterations occurring during development contributing to NB. We discuss current treatment regimens, present and future directions for identification of novel therapeutic metabolic targets, differentiation agents, as well as personalized combinatory therapies as potential approaches for improving survival and quality of life of children with NB.
Collapse
Affiliation(s)
- Lourdes Sainero-Alcolado
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, SE-17165, Stockholm, Sweden
| | - Tomas Sjöberg Bexelius
- Paediatric Oncology Unit, Astrid Lindgren's Children Hospital, SE-171 64 Solna, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Giuseppe Santopolo
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, SE-17165, Stockholm, Sweden
| | - Ye Yuan
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, SE-17165, Stockholm, Sweden
| | - Judit Liaño-Pons
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, SE-17165, Stockholm, Sweden
| | - Marie Arsenian-Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum B7, Karolinska Institutet, SE-17165, Stockholm, Sweden
- Lund University, Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, SE-223 81, Sweden
| |
Collapse
|
7
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
8
|
Pieniążek B, Cencelewicz K, Bździuch P, Młynarczyk Ł, Lejman M, Zawitkowska J, Derwich K. Neuroblastoma-A Review of Combination Immunotherapy. Int J Mol Sci 2024; 25:7730. [PMID: 39062971 PMCID: PMC11276848 DOI: 10.3390/ijms25147730] [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: 05/18/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor found in childhood and is responsible for 15% of deaths among children with cancer. Although multimodal therapies focused on surgery, chemotherapy, radiotherapy, and stem cell transplants have favorable results in many cases, the use of conventional therapies has probably reached the limit their possibility. Almost half of the patients with neuroblastoma belong to the high-risk group. Patients in this group require a combination of several therapeutic approaches. It has been shown that various immunotherapies combined with conventional methods can work synergistically. Due to the development of such therapeutic methods, we present combinations and forms of combining immunotherapy, focusing on their mechanisms and benefits but also their limitations and potential side effects.
Collapse
Affiliation(s)
- Barbara Pieniążek
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (B.P.); (K.C.); (P.B.)
| | - Katarzyna Cencelewicz
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (B.P.); (K.C.); (P.B.)
| | - Patrycja Bździuch
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (B.P.); (K.C.); (P.B.)
| | - Łukasz Młynarczyk
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (Ł.M.); (K.D.)
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantation, Medical University of Lublin, 20-093 Lublin, Poland
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (Ł.M.); (K.D.)
| |
Collapse
|
9
|
Xu S, Lan H, Huang C, Ge X, Zhu J. Mechanisms and emerging strategies for irinotecan-induced diarrhea. Eur J Pharmacol 2024; 974:176614. [PMID: 38677535 DOI: 10.1016/j.ejphar.2024.176614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Irinotecan (also known as CPT-11) is a topoisomerase I inhibitor first approved for clinical use as an anticancer agent in 1996. Over the past more than two decades, it has been widely used for combination regimens to treat various malignancies, especially in gastrointestinal and lung cancers. However, severe dose-limiting toxicities, especially gastrointestinal toxicity such as late-onset diarrhea, were frequently observed in irinotecan-based therapy, thus largely limiting the clinical application of this agent. Current knowledge regarding the pathogenesis of irinotecan-induced diarrhea is characterized by the complicated metabolism of irinotecan to its active metabolite SN-38 and inactive metabolite SN-38G. A series of enzymes and transporters were involved in these metabolic processes, including UGT1A1 and CYP3A4. Genetic polymorphisms of these metabolizing enzymes were significantly associated with the occurrence of irinotecan-induced diarrhea. Recent discoveries and progress made on the detailed mechanisms enable the identification of potential biomarkers for predicting diarrhea and as such guiding the proper patient selection with a better range of tolerant dosages. In this review, we introduce the metabolic process of irinotecan and describe the pathogenic mechanisms underlying irinotecan-induced diarrhea. Based on the mechanisms, we further outline the potential biomarkers for predicting the severity of diarrhea. Finally, based on the current experimental evidence in preclinical and clinical studies, we discuss and prospect the current and emerging strategies for the prevention of irinotecan-induced diarrhea.
Collapse
Affiliation(s)
- Shengkun Xu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Chengyi Huang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Xingnan Ge
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
10
|
Corbacioglu S, Lode H, Ellinger S, Zeman F, Suttorp M, Escherich G, Bochennek K, Gruhn B, Lang P, Rohde M, Debatin KM, Steinbach D, Beilken A, Ladenstein R, Spachtholz R, Heiss P, Hellwig D, Tröger A, Koller M, Menhart K, Riemenschneider MJ, Zoubaa S, Kietz S, Jakob M, Sommer G, Heise T, Hundsdörfer P, Kühnle I, Dilloo D, Schönberger S, Schwabe G, von Luettichau I, Graf N, Schlegel PG, Frühwald M, Jorch N, Paulussen M, Schneider DT, Metzler M, Leipold A, Nathrath M, Imschweiler T, Christiansen H, Schmid I, Crazzolara R, Niktoreh N, Cario G, Faber J, Demmert M, Babor F, Fröhlich B, Bielack S, Bernig T, Greil J, Eggert A, Simon T, Foell J. Irinotecan and temozolomide in combination with dasatinib and rapamycin versus irinotecan and temozolomide for patients with relapsed or refractory neuroblastoma (RIST-rNB-2011): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol 2024; 25:922-932. [PMID: 38936379 DOI: 10.1016/s1470-2045(24)00202-x] [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: 12/18/2023] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Neuroblastoma is the most common extracranial solid tumour in children. Relapsed or refractory neuroblastoma is associated with a poor outcome. We assessed the combination of irinotecan-temozolomide and dasatinib-rapamycin (RIST) in patients with relapsed or refractory neuroblastoma. METHODS The multicentre, open-label, randomised, controlled, phase 2, RIST-rNB-2011 trial recruited from 40 paediatric oncology centres in Germany and Austria. Patients aged 1-25 years with high-risk relapsed (defined as recurrence of all stage IV and MYCN amplification stages, after response to treatment) or refractory (progressive disease during primary treatment) neuroblastoma, with Lansky and Karnofsky performance status at least 50%, were assigned (1:1) to RIST (RIST group) or irinotecan-temozolomide (control group) by block randomisation, stratified by MYCN status. We compared RIST (oral rapamycin [loading 3 mg/m2 on day 1, maintenance 1 mg/m2 on days 2-4] and oral dasatinib [2 mg/kg per day] for 4 days with 3 days off, followed by intravenous irinotecan [50 mg/m2 per day] and oral temozolomide [150 mg/m2 per day] for 5 days with 2 days off; one course each of rapamycin-dasatinib and irinotecan-temozolomide for four cycles over 8 weeks, then two courses of rapamycin-dasatinib followed by one course of irinotecan-temozolomide for 12 weeks) with irinotecan-temozolomide alone (with identical dosing as experimental group). The primary endpoint of progression-free survival was analysed in all eligible patients who received at least one course of therapy. The safety population consisted of all patients who received at least one course of therapy and had at least one post-baseline safety assessment. This trial is registered at ClinicalTrials.gov, NCT01467986, and is closed to accrual. FINDINGS Between Aug 26, 2013, and Sept 21, 2020, 129 patients were randomly assigned to the RIST group (n=63) or control group (n=66). Median age was 5·4 years (IQR 3·7-8·1). 124 patients (78 [63%] male and 46 [37%] female) were included in the efficacy analysis. At a median follow-up of 72 months (IQR 31-88), the median progression-free survival was 11 months (95% CI 7-17) in the RIST group and 5 months (2-8) in the control group (hazard ratio 0·62, one-sided 90% CI 0·81; p=0·019). Median progression-free survival in patients with amplified MYCN (n=48) was 6 months (95% CI 4-24) in the RIST group versus 2 months (2-5) in the control group (HR 0·45 [95% CI 0·24-0·84], p=0·012); median progression-free survival in patients without amplified MYCN (n=76) was 14 months (95% CI 9-7) in the RIST group versus 8 months (4-15) in the control group (HR 0·84 [95% CI 0·51-1·38], p=0·49). The most common grade 3 or worse adverse events were neutropenia (54 [81%] of 67 patients given RIST vs 49 [82%] of 60 patients given control), thrombocytopenia (45 [67%] vs 41 [68%]), and anaemia (39 [58%] vs 38 [63%]). Nine serious treatment-related adverse events were reported (five patients given control and four patients given RIST). There were no treatment-related deaths in the control group and one in the RIST group (multiorgan failure). INTERPRETATION RIST-rNB-2011 demonstrated that targeting of MYCN-amplified relapsed or refractory neuroblastoma with a pathway-directed metronomic combination of a multkinase inhibitor and an mTOR inhibitor can improve progression-free survival and overall survival. This exclusive efficacy in MYCN-amplified, relapsed neuroblastoma warrants further investigation in the first-line setting. FUNDING Deutsche Krebshilfe.
Collapse
Affiliation(s)
- Selim Corbacioglu
- University Medical Center Regensburg, Regensburg, Germany; Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation,University Medical Center Regensburg, Regensburg, Germany.
| | - Holger Lode
- University Medicine Greifswald, Greifswald, Germany
| | | | - Florian Zeman
- University Medical Center Regensburg, Regensburg, Germany
| | - Meinolf Suttorp
- Medical Faculty, Technical University Dresden, Dresden, Germany
| | | | | | - Bernd Gruhn
- University Medical Center Jena, Jena, Germany
| | - Peter Lang
- University Medical Center Tuebingen, Tuebingen, Germany
| | - Marius Rohde
- University Medical Center Giessen, Giessen, Germany
| | | | | | | | - Ruth Ladenstein
- St Anna Children's Cancer Research Institute, University Medical Center, Vienna, Austria
| | | | - Peter Heiss
- University Medical Center Regensburg, Regensburg, Germany
| | - Dirk Hellwig
- University Medical Center Regensburg, Regensburg, Germany
| | - Anja Tröger
- University Medical Center Regensburg, Regensburg, Germany
| | - Michael Koller
- University Medical Center Regensburg, Regensburg, Germany
| | - Karin Menhart
- University Medical Center Regensburg, Regensburg, Germany
| | | | - Saida Zoubaa
- University Medical Center Regensburg, Regensburg, Germany
| | - Silke Kietz
- University Medical Center Regensburg, Regensburg, Germany
| | - Marcus Jakob
- University Medical Center Regensburg, Regensburg, Germany
| | - Gunhild Sommer
- University Medical Center Regensburg, Regensburg, Germany
| | - Tilman Heise
- University Medical Center Regensburg, Regensburg, Germany
| | | | - Ingrid Kühnle
- University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | - Norbert Jorch
- University Medical Center Bielefeld, Bielefeld, Germany
| | | | | | | | | | | | | | | | - Irene Schmid
- Ludwig Maximilians University Munich, Munich, Germany
| | | | | | | | - Joerg Faber
- University Medical Center Mainz, Mainz, Germany
| | | | - Florian Babor
- University Medical Center Düsseldorf, Düsseldorf, Germany
| | | | | | | | - Johann Greil
- University Medical Center Heidelberg, Heidelberg, Germany
| | | | | | - Juergen Foell
- University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
11
|
Li H, Zhang P, Yuan X, Peng S, Yang X, Li Y, Shen Z, Bai J. Targeted drug-loaded peptides induce tumor cell apoptosis and immunomodulation to increase antitumor efficacy. BIOMATERIALS ADVANCES 2024; 160:213852. [PMID: 38636118 DOI: 10.1016/j.bioadv.2024.213852] [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: 11/22/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/20/2024]
Abstract
Immunotherapy is an emerging approach for the treatment of solid tumors. Although chemotherapy is generally considered immunosuppressive, specific chemotherapeutic agents can induce tumor immunity. In this study, we developed a targeted, acid-sensitive peptide nanoparticle (DT/Pep1) to deliver doxorubicin (DOX) and triptolide (TPL) to breast cancer cells via the enhanced permeability and retention (EPR) effect and the breast cancer-targeting effect of peptide D8. Compared with administration of the free drugs, treatment with the DT/Pep1 system increased the accumulation of DOX and TPL at the tumor site and achieved deeper penetration into the tumor tissue. In an acidic environment, DT/Pep1 transformed from spherical nanoparticles to aggregates with a high aspect ratio, which successfully extended the retention of the drugs in the tumor cells and bolstered the anticancer effect. In both in vivo and in vitro experiments, DT/Pep1 effectively blocked the cell cycle and induced apoptosis. Importantly, the DT/Pep1 system efficiently suppressed tumor development in mice bearing 4T1 tumors while simultaneously promoting immune system activation. Thus, the results of this study provide a system for breast cancer therapy and offer a novel and promising platform for peptide nanocarrier-based drug delivery.
Collapse
Affiliation(s)
- Hongjie Li
- School of Medical Sciences, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, China
| | - Peirong Zhang
- School of Medical Sciences, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, China
| | - Xiaomeng Yuan
- School of Bioscience and Technology, Shandong Second Medical University, Weifang 261053, China
| | - Shan Peng
- School of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Xingyue Yang
- School of Bioscience and Technology, Shandong Second Medical University, Weifang 261053, China
| | - Yuxia Li
- School of Medical Sciences, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang 261053, China
| | - Zhen Shen
- Clinical laboratory, Affiliated Hospital of Shandong Second Medical University, Weifang 261053, China
| | - Jingkun Bai
- School of Bioscience and Technology, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
12
|
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
|
13
|
Polychronopoulos PA, Bedoya-Reina OC, Johnsen JI. The Neuroblastoma Microenvironment, Heterogeneity and Immunotherapeutic Approaches. Cancers (Basel) 2024; 16:1863. [PMID: 38791942 PMCID: PMC11119056 DOI: 10.3390/cancers16101863] [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: 04/08/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Neuroblastoma is a peripheral nervous system tumor that almost exclusively occurs in young children. Although intensified treatment modalities have led to increased patient survival, the prognosis for patients with high-risk disease is still around 50%, signifying neuroblastoma as a leading cause of cancer-related deaths in children. Neuroblastoma is an embryonal tumor and is shaped by its origin from cells within the neural crest. Hence, neuroblastoma usually presents with a low mutational burden and is, in the majority of cases, driven by epigenetically deregulated transcription networks. The recent development of Omic techniques has given us detailed knowledge of neuroblastoma evolution, heterogeneity, and plasticity, as well as intra- and intercellular molecular communication networks within the neuroblastoma microenvironment. Here, we discuss the potential of these recent discoveries with emphasis on new treatment modalities, including immunotherapies which hold promise for better future treatment regimens.
Collapse
Affiliation(s)
- Panagiotis Alkinoos Polychronopoulos
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
| | - Oscar C. Bedoya-Reina
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
- School of Medical Sciences, Örebro University, 70182 Örebro, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, 11883 Stockholm, Sweden; (P.A.P.); (O.C.B.-R.)
| |
Collapse
|
14
|
Raiser P, Schleiermacher G, Gambart M, Dumont B, Defachelles AS, Thebaud E, Tandonnet J, Pasqualini C, Proust S, Entz-Werle N, Aerts I, Ndounga-Diakou LA, Petit A, Puiseux C, Khanfar C, Rouger J, Mansuy L, Benadiba J, Millot F, Pluchart C, Laghouati S, Geoerger B, Vassal G, Valteau-Couanet D, Berlanga P. Chemo-immunotherapy with dinutuximab beta in patients with relapsed/progressive high-risk neuroblastoma: does chemotherapy backbone matter? Eur J Cancer 2024; 202:114001. [PMID: 38489858 DOI: 10.1016/j.ejca.2024.114001] [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: 01/16/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Addition of anti-GD2 antibodies to temozolomide-based chemotherapy has demonstrated increased antitumor activity and progression-free survival in patients with relapsed/progressive high-risk neuroblastoma. However, chemo-immunotherapy is not yet approved for this indication. This study presents the chemo-immunotherapy experience in patients with relapsed/progressive high-risk neuroblastoma treated within the off-label use program of the Neuroblastoma Committee of the French Society of Pediatric Oncology (SFCE). METHODS Dinutuximab beta (dB) was administered alongside temozolomide-topotecan (TOTEM) or temozolomide-irinotecan (TEMIRI) at first disease relapse/progression or topotecan-cyclophosphamide (TopoCyclo) at further relapse/progression. Real-world data on demographics, treatment, antitumor activity and safety was collected from all patients after inclusion in SACHA-France (NCT04477681), a prospective national registry, which documents safety and efficacy data on innovative anticancer therapies prescribed to patients ≤ 25 years old as compassionate or off-label use. RESULTS Between February 2021 and July 2023, 39 patients with confirmed relapsed/progressive high-risk neuroblastoma (median age 6 years, range 1-24) were treated with dB+TopoCyclo (n = 24) or dB+TOTEM/TEMIRI (n = 15) across 17 centers. In total, 163 chemo-immunotherapy cycles were administered, main toxicities were mild or moderate, with higher incidence of hematological adverse drug reactions with dB+TopoCyclo than dB+TOTEM/TEMIRI. Objective response rate was 42% for dB+TopoCyclo (CI95% 22-63%) and 40% for dB+TOTEM/TEMIRI (CI95% 16-68%). CONCLUSION Similar objective response rates for dB+TopoCyclo and dB+TOTEM/TEMIRI in patients with relapsed/progressive high-risk neuroblastoma emphasize the importance of chemo-immunotherapy, irrespective of the chemotherapy backbone.
Collapse
Affiliation(s)
- Patricia Raiser
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Gudrun Schleiermacher
- RTOP (Recherche Translationelle en Oncologie Pediatrique), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France; SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France
| | - Marion Gambart
- Department of Pediatric Oncology, Toulouse University Hospital, 31300 Toulouse, France
| | - Benoit Dumont
- Department of Pediatric Oncology, Institut d'Hématologie et d'Oncologie Pédiatrique/Centre Léon Bérard, 69008 Lyon, France
| | | | - Estelle Thebaud
- Pediatric Immuno-Hemato-Oncology Unit, CHU Nantes, 44000 Nantes, France
| | - Julie Tandonnet
- Pediatric Hematology-Oncology Department, Centre Hospitalier Universitaire (CHU), 33000 Bordeaux, France
| | - Claudia Pasqualini
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Stéphanie Proust
- Department of Pediatric Oncology, University Hospital, 49100 Angers, France
| | - Natacha Entz-Werle
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67200 Strasbourg, France
| | - Isabelle Aerts
- RTOP (Recherche Translationelle en Oncologie Pediatrique), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France; SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France
| | - Lee A Ndounga-Diakou
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Arnaud Petit
- Department of Pediatric Hematology and Oncology, Hôpital Armand Trousseau, 75012 Paris, France
| | - Chloe Puiseux
- Department of Pediatric Hemato-Oncology, University Hospital of Rennes, 35200 Rennes, France
| | - Camille Khanfar
- Department of Pediatric Oncology, CHU Amiens Picardie, 80054 Amiens, France
| | - Jeremie Rouger
- Department of Pediatric Hematology and Oncology, University Hospital of Caen, 14000 Caen, France
| | - Ludovic Mansuy
- Department of Pediatric Hematology-Oncology, Children's Hospital of Brabois, 54500 Vandoeuvre Les Nancy, France
| | - Joy Benadiba
- Department of Hemato-Oncology Pediatric, Nice University Hospital, 06000 Nice, France
| | - Frédéric Millot
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France
| | - Claire Pluchart
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Reims, 51100 Reims, France
| | - Salim Laghouati
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France; INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Gilles Vassal
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Dominique Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Pablo Berlanga
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France.
| |
Collapse
|
15
|
Conte B, Casey DL, Tringale KR, Honeyman J, Narayan NJC, LaQuaglia MP, Gerstle JT, Modak S, Kushner BH, Kramer K, Wolden SL. Intraoperative Radiation Therapy for Relapsed or Refractory High-Risk Neuroblastoma: A 27-Year Experience. Pract Radiat Oncol 2024; 14:e226-e232. [PMID: 38310488 DOI: 10.1016/j.prro.2023.12.008] [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: 08/31/2023] [Revised: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 02/05/2024]
Abstract
PURPOSE To evaluate outcomes after intraoperative radiation therapy (IORT) in high-risk neuroblastoma (NB), including local control, overall survival, and toxicity. METHODS AND MATERIALS This was a single institution retrospective study of 92 pediatric patients with NB treated with IORT from 1995 to 2022. Each IORT application was considered a separate event for a total of 110 sites treated. Local failure was calculated using the cumulative incidence function and survival by Kaplan-Meier method from the day of surgery. RESULTS All patients had high-risk relapsed or treatment refractory disease. Median age was 6 years (range, 2-34 years). Median follow-up for all patients and surviving patients was 16 months and 4 years, respectively. All patients previously received chemotherapy, 93% had prior external beam radiation therapy to the site of IORT (median dose, 21.6 Gy; range, 10-36 Gy), and 94% had a prior surgery for tumor resection. The median IORT dose was 12 Gy (range, 8-18 Gy) and median area treated was 18 cm2 (range, 2.5-60 cm2). The cumulative incidence of local failure was 23% at 2 years and 29% at 5 years. The overall survival (OS) was 44% at 2 years and 29% at 5 years. Local failure after IORT was associated with worse OS (hazard ratio, 1.74; 95% CI, 1.07-2.84; P = .0267). Toxicity from IORT was rare, with postoperative complications likely related to IORT seen in 7 (8%) patients. CONCLUSIONS Our study represents the largest, most recent analysis of the efficacy and safety of IORT in patients with relapsed or refractory NB. Less than one-third of patients failed locally at 5 years, and achieving local control affected overall survival. Minimal toxicities directly related to IORT were observed. Overall, IORT is an effective and safe technique to achieve local control in high-risk relapsed or refractory neuroblastoma.
Collapse
Affiliation(s)
- Brianna Conte
- University of Miami Miller School of Medicine, Miami, Florida.
| | - Dana L Casey
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Kathryn R Tringale
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua Honeyman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicole J C Narayan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael P LaQuaglia
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Ted Gerstle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shakeel Modak
- Department of Pediatrics, Neuroblastoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian H Kushner
- Department of Pediatrics, Neuroblastoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kim Kramer
- Department of Pediatrics, Neuroblastoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
16
|
Gorostegui M, Muñoz JP, Perez-Jaume S, Simao-Rafael M, Larrosa C, Garraus M, Salvador N, Lavarino C, Krauel L, Mañe S, Castañeda A, Mora J. Management of High-Risk Neuroblastoma with Soft-Tissue-Only Disease in the Era of Anti-GD2 Immunotherapy. Cancers (Basel) 2024; 16:1735. [PMID: 38730688 PMCID: PMC11083939 DOI: 10.3390/cancers16091735] [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: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma presents with two patterns of disease: locoregional or systemic. The poor prognostic risk factors of locoregional neuroblastoma (LR-NB) include age, MYCN or MDM2-CDK4 amplification, 11q, histology, diploidy with ALK or TERT mutations, and ATRX aberrations. Anti-GD2 immunotherapy has significantly improved the outcome of high-risk (HR) NB and is mostly effective against osteomedullary minimal residual disease (MRD), but less so against soft tissue disease. The question is whether adding anti-GD2 monoclonal antibodies (mAbs) benefits patients with HR-NB compounded by only soft tissue. We reviewed 31 patients treated at SJD for HR-NB with no osteomedullary involvement at diagnosis. All tumors had molecular genetic features of HR-NB. The outcome after first-line treatment showed 25 (80.6%) patients achieving CR. Thirteen patients remain in continued CR, median follow-up 3.9 years. We analyzed whether adding anti-GD2 immunotherapy to first-line treatment had any prognostic significance. The EFS analysis using Cox models showed a HR of 0.20, p = 0.0054, and an 80% decrease in the risk of relapse in patients treated with anti-GD2 immunotherapy in the first line. Neither EFS nor OS were significantly different by CR status after first-line treatment. In conclusion, adding treatment with anti-GD2 mAbs at the stage of MRD helps prevent relapse that unequivocally portends poor survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (M.G.); (J.P.M.); (M.S.-R.); (C.L.); (M.G.); (N.S.); (C.L.); (L.K.); (S.M.); (A.C.)
| |
Collapse
|
17
|
Zebertavage L, Schopf A, Nielsen M, Matthews J, Erbe AK, Aiken TJ, Katz S, Sun C, Witt CM, Rakhmilevich AL, Sondel PM. Evaluation of a Combinatorial Immunotherapy Regimen That Can Cure Mice Bearing MYCN-Driven High-Risk Neuroblastoma That Resists Current Clinical Therapy. J Clin Med 2024; 13:2561. [PMID: 38731089 PMCID: PMC11084214 DOI: 10.3390/jcm13092561] [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: 02/22/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Incorporating GD2-targeting monoclonal antibody into post-consolidation maintenance therapy has improved survival for children with high-risk neuroblastoma. However, ~50% of patients do not respond to, or relapse following, initial treatment. Here, we evaluated additional anti-GD2-based immunotherapy to better treat high-risk neuroblastoma in mice to develop a regimen for patients with therapy-resistant neuroblastoma. Methods: We determined the components of a combined regimen needed to cure mice of established MYCN-amplified, GD2-expressing, murine 9464D-GD2 neuroblastomas. Results: First, we demonstrate that 9464D-GD2 is nonresponsive to a preferred salvage regimen: anti-GD2 with temozolomide and irinotecan. Second, we have previously shown that adding agonist anti-CD40 mAb and CpG to a regimen of radiotherapy, anti-GD2/IL2 immunocytokine and anti-CTLA-4, cured a substantial fraction of mice bearing small 9464D-GD2 tumors; here, we further characterize this regimen by showing that radiotherapy and hu14.18-IL2 are necessary components, while anti-CTLA-4, anti-CD40, or CpG can individually be removed, and CpG and anti-CTLA-4 can be removed together, while maintaining efficacy. Conclusions: We have developed and characterized a regimen that can cure mice of a high-risk neuroblastoma that is refractory to the current clinical regimen for relapsed/refractory disease. Ongoing preclinical work is directed towards ways to potentially translate these findings to a regimen appropriate for clinical testing.
Collapse
Affiliation(s)
- Lauren Zebertavage
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Allison Schopf
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Megan Nielsen
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Joel Matthews
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Taylor J. Aiken
- Department of Surgery, University of Wisconsin, Madison, WI 53705, USA;
| | - Sydney Katz
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Claire Sun
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Cole M. Witt
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Alexander L. Rakhmilevich
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA (A.S.); (M.N.); (J.M.); (A.K.E.); (S.K.); (C.S.); (C.M.W.); (A.L.R.)
- Department of Pediatrics, University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
18
|
Mora J, Modak S, Kinsey J, Ragsdale CE, Lazarus HM. GM-CSF, G-CSF or no cytokine therapy with anti-GD2 immunotherapy for high-risk neuroblastoma. Int J Cancer 2024; 154:1340-1364. [PMID: 38108214 DOI: 10.1002/ijc.34815] [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: 06/14/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
Colony-stimulating factors have been shown to improve anti-disialoganglioside 2 (anti-GD2) monoclonal antibody response in high-risk neuroblastoma by enhancing antibody-dependent cell-mediated cytotoxicity (ADCC). A substantial amount of research has focused on recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF) as an adjuvant to anti-GD2 monoclonal antibodies. There may be a disparity in care among patients as access to GM-CSF therapy and anti-GD2 monoclonal antibodies is not uniform. Only select countries have approved these agents for use, and even with regulatory approvals, access to these agents can be complex and cost prohibitive. This comprehensive review summarizes clinical data regarding efficacy and safety of GM-CSF, recombinant human granulocyte colony-stimulating factor (G-CSF) or no cytokine in combination with anti-GD2 monoclonal antibodies (ie, dinutuximab, dinutuximab beta or naxitamab) for immunotherapy of patients with high-risk neuroblastoma. A substantial body of clinical data support the immunotherapy combination of anti-GD2 monoclonal antibodies and GM-CSF. In contrast, clinical data supporting the use of G-CSF are limited. No formal comparison between GM-CSF, G-CSF and no cytokine has been identified. The treatment of high-risk neuroblastoma with anti-GD2 therapy plus GM-CSF is well established. Suboptimal efficacy outcomes with G-CSF raise concerns about its suitability as an alternative to GM-CSF as an adjuvant in immunotherapy for patients with high-risk neuroblastoma. While programs exist to facilitate obtaining GM-CSF and anti-GD2 monoclonal antibodies in regions where they are not commercially available, continued work is needed to ensure equitable therapeutic options are available globally.
Collapse
Affiliation(s)
- Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Shakeel Modak
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joyce Kinsey
- Partner Therapeutics, Inc, Lexington, Massachusetts, USA
| | | | | |
Collapse
|
19
|
Lee WG, Kim ES. Precision Oncology in Pediatric Cancer Surgery. Surg Oncol Clin N Am 2024; 33:409-446. [PMID: 38401917 DOI: 10.1016/j.soc.2023.12.008] [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] [Indexed: 02/26/2024]
Abstract
Pediatric precision oncology has provided a greater understanding of the wide range of molecular alterations in difficult-to-treat or rare tumors with the aims of increasing survival as well as decreasing toxicity and morbidity from current cytotoxic therapies. In this article, the authors discuss the current state of pediatric precision oncology which has increased access to novel targeted therapies while also providing a framework for clinical implementation in this unique population. The authors evaluate the targetable mutations currently under investigation-with a focus on pediatric solid tumors-and discuss the key surgical implications associated with novel targeted therapies.
Collapse
Affiliation(s)
- William G Lee
- Department of Surgery, Cedars-Sinai Medical Center, 116 North Robertson Boulevard, Suite PACT 700, Los Angeles, CA 90048, USA. https://twitter.com/william_ghh_lee
| | - Eugene S Kim
- Division of Pediatric Surgery, Department of Surgery, Cedars-Sinai Medical Center, 116 North Robertson Boulevard, Suite PACT 700, Los Angeles, CA 90048, USA.
| |
Collapse
|
20
|
Chaix J, Schleiermacher G, Corradini N, André N, Thebaud E, Gambart M, Defachelles AS, Entz-Werle N, Chastagner P, De Carli É, Ducassou S, Landman-Parker J, Adam-de-Beaumais T, Larive A, Michiels S, Vassal G, Valteau-Couanet D, Geoerger B, Berlanga P. Clinical trial inclusion in patients with relapsed/refractory neuroblastoma following the European Precision Cancer Medicine trial MAPPYACTS. Eur J Cancer 2024; 201:113923. [PMID: 38377775 DOI: 10.1016/j.ejca.2024.113923] [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: 11/11/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024]
Abstract
INTRODUCTION Despite poor survival for patients with relapsed or refractory neuroblastoma, only 10-16% of patients are reported to be included in early phase trials. This study aimed to explore the impact of molecular profiling within the prospective precision cancer medicine trial MAPPYACTS (NCT02613962) on subsequent early phase trial recruitment and treatment by matched targeted therapies in this population. METHODS AND MATERIALS Clinical data from all French patients with relapsed/refractory neuroblastoma enrolled in MAPPYACTS were analyzed for subsequent matched/non-matched targeted treatment based on clinical tumor board (CMTB) recommendations. RESULTS From 93 patients with neuroblastoma included in French centers, 78 (84%) underwent whole exome and RNA sequencing and were discussed in the CMTB. Higher rate of successful sequencing analysis was observed in patients with relapsed disease compared to those with refractory disease (p = 0.0002). Among the 50 patients that presented with a new disease relapse/progression after the CMTB recommendations, 35 patients (70%) had at least one actionable alteration identified on the tumor at the time of relapse. Eighteen patients (36%) were included in an early phase clinical trial, 11 of these with a matched agent, 7 with a non-matched treatment; 13 patients were included in the AcSé ESMART trial. Five patients (10%) received a matched targeted therapy outside a clinical trial. CONCLUSION Patients with neuroblastoma in the European MAPPYACTS trial were more likely to be included in early phase trials compared to previous reports. Early deep sequencing at first treatment failure, comprehensive therapeutic discussions in molecular tumor boards and innovative trials like AcSé -ESMART improve access to innovative therapies for patients with relapsed/refractory neuroblastoma. CLINICAL TRIAL REGISTRATION NCT02613962.
Collapse
Affiliation(s)
- Jordane Chaix
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Gudrun Schleiermacher
- INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Research Center, PSL Research University, Institut Curie, Paris, France; SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), Institut Curie, PSL Research University, Paris, France
| | - Nadège Corradini
- Department of Pediatric Oncology, Institut d'Hématologie et d'Oncologie Pédiatrique/Centre Léon Bérard, Lyon, France
| | - Nicolas André
- Department of Pediatric Hematology and Oncology, Hôpital de La Timone, AP-HM, Marseille, France; UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université U105, Marseille Cancer Research Center (CRCM), Marseille, France
| | - Estelle Thebaud
- Department of Pediatric Oncology, Centre Hospitalier Universitaire, Nantes, France
| | - Marion Gambart
- Department of Pediatric Oncology, Centre Hospitalier Universitaire, Toulouse, France
| | | | - Natacha Entz-Werle
- Department of Pediatric Oncology, Hospices Civils de Strasbourg, Strasbourg, France
| | - Pascal Chastagner
- Department of Pediatric Oncology, Centre Hospitalier Universitaire, Vandoeuvre les Nancy, France
| | - Émilie De Carli
- Department of Pediatric Oncology, Centre Hospitalier Universitaire, Angers, France
| | - Stéphane Ducassou
- Department of Pediatric Oncology, Centre Hospitalier Universitaire, Bordeaux, France
| | | | - Tiphaine Adam-de-Beaumais
- Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Alicia Larive
- Biostatistics and Epidemiology Office, Gustave Roussy Cancer Campus, INSERM U1018, CESP, Université Paris-Saclay, Villejuif, France
| | - Stefan Michiels
- Biostatistics and Epidemiology Office, Gustave Roussy Cancer Campus, INSERM U1018, CESP, Université Paris-Saclay, Villejuif, France
| | - Gilles Vassal
- Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Dominique Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France; INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Pablo Berlanga
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
21
|
Philippova J, Shevchenko J, Sennikov S. GD2-targeting therapy: a comparative analysis of approaches and promising directions. Front Immunol 2024; 15:1371345. [PMID: 38558810 PMCID: PMC10979305 DOI: 10.3389/fimmu.2024.1371345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Disialoganglioside GD2 is a promising target for immunotherapy with expression primarily restricted to neuroectodermal and epithelial tumor cells. Although its role in the maintenance and repair of neural tissue is well-established, its functions during normal organism development remain understudied. Meanwhile, studies have shown that GD2 plays an important role in tumorigenesis. Its functions include proliferation, invasion, motility, and metastasis, and its high expression and ability to transform the tumor microenvironment may be associated with a malignant phenotype. Structurally, GD2 is a glycosphingolipid that is stably expressed on the surface of tumor cells, making it a suitable candidate for targeting by antibodies or chimeric antigen receptors. Based on mouse monoclonal antibodies, chimeric and humanized antibodies and their combinations with cytokines, toxins, drugs, radionuclides, nanoparticles as well as chimeric antigen receptor have been developed. Furthermore, vaccines and photoimmunotherapy are being used to treat GD2-positive tumors, and GD2 aptamers can be used for targeting. In the field of cell therapy, allogeneic immunocompetent cells are also being utilized to enhance GD2 therapy. Efforts are currently being made to optimize the chimeric antigen receptor by modifying its design or by transducing not only αβ T cells, but also γδ T cells, NK cells, NKT cells, and macrophages. In addition, immunotherapy can combine both diagnostic and therapeutic methods, allowing for early detection of disease and minimal residual disease. This review discusses each immunotherapy method and strategy, its advantages and disadvantages, and highlights future directions for GD2 therapy.
Collapse
Affiliation(s)
| | | | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
22
|
Fiz F, Piticchio T, Bottoni G, Sorrentino S, Fragola M, Livellara V, Trimboli P, Piccardo A. Incidence of subclinical and overt hypothyroidism in children treated with [131I]mIBG: a systematic review and meta-analysis. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2024; 68:40-47. [PMID: 38319674 DOI: 10.23736/s1824-4785.24.03552-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Treatment with [131I]mIBG is commonly used in pediatric metastatic neuroblastoma (NB); however, unbound [131I]I might be taken up by the thyroid, causing hypothyroidism. To prevent this occurrence, thyroid blockade with iodine salts is commonly used; despite this precaution, thyroid dysfunction still occurs. This review and meta-analysis aim to clarify the mean frequency of hypothyroidism in children with NB treated with [131I]mIBG and to investigate the possible causes. EVIDENCE ACQUISITION The literature was searched for English-language scientific manuscripts describing the incidence of TSH elevation and overt hypothyroidism in children with NB treated with [131I]mIBG. Preclinical studies, small-case series, and reviews were excluded. A proportion meta-analysis was conducted to test the influence of potentially relevant factors (type and duration of thyroid blockade, year of the study, sample size) on the incidence of TSH elevation/overt hypothyroidism. EVIDENCE SYNTHESIS Eleven studies were included. The pooled percentage of TSH elevation was 0.41 (95% CI: 0.27-0.55); the duration of the thyroid blockade (P=0.004) was inversely correlated with the incidence of TSH elevation. Moreover, a TSH increase was more common in patients treated with potassium iodide (KI) alone than in those managed with a multi-drug thyroid blockade (P<0.001). The pooled percentage of children requiring hormone replacement therapy was 0.33 (95% CI: 0.16-0.49). As in the case of TSH elevation, a longer duration of the thyroid blockade (P=0.006) and a multi-pronged approach (P<0.001) were associated with a lower incidence of overt hypothyroidism. CONCLUSIONS Hypothyroidism appears to occur frequently in children treated with [131I]mIBG, which should be monitored closely after the radionuclide treatment to start hormone replacement therapy as soon as needed. The duration, as well as the type of thyroid blockade, seem to influence the incidence of hypothyroidism; however, more data from prospective evaluations are needed.
Collapse
Affiliation(s)
- Francesco Fiz
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Genoa, Italy
- Department of Nuclear Medicine and Clinical Molecular Imaging, Tübingen University Hospital, Tübingen, Germany
| | - Tommaso Piticchio
- Section of Endocrinology, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, Catania, Italy
| | - Gianluca Bottoni
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Genoa, Italy
| | | | - Martina Fragola
- Biostatistics Unit, Scientific Directorate, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Pierpaolo Trimboli
- Clinic of Endocrinology and Diabetology, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, University of Italian Switzerland (USI), Lugano, Switzerland
| | - Arnoldo Piccardo
- Department of Nuclear Medicine, E.O. Ospedali Galliera, Genoa, Italy -
| |
Collapse
|
23
|
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
|
24
|
Hogarty MD, Ziegler DS, Franson A, Chi YY, Tsao-Wei D, Liu K, Vemu R, Gerner EW, Bruckheimer E, Shamirian A, Hasenauer B, Balis FM, Groshen S, Norris MD, Haber M, Park JR, Matthay KK, Marachelian A. Phase 1 study of high-dose DFMO, celecoxib, cyclophosphamide and topotecan for patients with relapsed neuroblastoma: a New Approaches to Neuroblastoma Therapy trial. Br J Cancer 2024; 130:788-797. [PMID: 38200233 PMCID: PMC10912730 DOI: 10.1038/s41416-023-02525-2] [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: 10/10/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND MYC genes regulate ornithine decarboxylase (Odc) to increase intratumoral polyamines. We conducted a Phase I trial [NCT02030964] to determine the maximum tolerated dose (MTD) of DFMO, an Odc inhibitor, with celecoxib, cyclophosphamide and topotecan. METHODS Patients 2-30 years of age with relapsed/refractory high-risk neuroblastoma received oral DFMO at doses up to 9000 mg/m2/day, with celecoxib (500 mg/m2 daily), cyclophosphamide (250 mg/m2/day) and topotecan (0.75 mg/m2/day) IV for 5 days, for up to one year with G-CSF support. RESULTS Twenty-four patients (median age, 6.8 years) received 136 courses. Slow platelet recovery with 21-day courses (dose-levels 1 and 2) led to subsequent dose-levels using 28-day courses (dose-levels 2a-4a). There were three course-1 dose-limiting toxicities (DLTs; hematologic; anorexia; transaminases), and 23 serious adverse events (78% fever-related). Five patients (21%) completed 1-year of therapy. Nine stopped for PD, 2 for DLT, 8 by choice. Best overall response included two PR and four MR. Median time-to-progression was 19.8 months, and 3 patients remained progression-free at >4 years without receiving additional therapy. The MTD of DFMO with this regimen was 6750 mg/m2/day. CONCLUSION High-dose DFMO is tolerable when added to chemotherapy in heavily pre-treated patients. A randomized Phase 2 trial of DFMO added to chemoimmunotherapy is ongoing [NCT03794349].
Collapse
Affiliation(s)
- Michael D Hogarty
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Andrea Franson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yueh-Yun Chi
- Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Denice Tsao-Wei
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kangning Liu
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rohan Vemu
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Anasheh Shamirian
- Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Beth Hasenauer
- Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Frank M Balis
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Groshen
- Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Julie R Park
- St. Jude Children's Research Hospital, University of Tennessee, Memphis, TN, USA
| | - Katherine K Matthay
- UCSF Benioff Children's Hospital, UCSF School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Araz Marachelian
- Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
25
|
Yan H, Wang P, Yang F, Cheng W, Chen C, Zhai B, Zhou Y. Anticancer therapy-induced adverse drug reactions in children and preventive and control measures. Front Pharmacol 2024; 15:1329220. [PMID: 38425652 PMCID: PMC10902428 DOI: 10.3389/fphar.2024.1329220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
In recent years, considerable achievements have been made in pediatric oncology with the innovation and development of antitumor drugs. However, compared to adults, children as a special group have not yet matured fully in terms of liver and kidney function. Moreover, pediatric patients are prone to more adverse drug reactions (ADRs) from the accumulation of antineoplastic drugs due to their smaller body size and larger body surface area. Chemotherapy-related ADRs have become a non-negligible factor that affects cancer remission. To date, studies on ADRs in pediatric cancer patients have emerged internationally, but few systematic summaries are available. Here, we reviewed the various systemic ADRs associated with antitumor drugs in children and adolescent patients, as well as the advances in strategies to cope with ADRs, which consisted of neurotoxicity, hematological toxicity, cardiotoxicity, ADRs of the respiratory system and gastrointestinal system and urinary system, ADRs of the skin and its adnexa, allergic reactions, and other ADRs. For clinicians and researchers, understanding the causes, symptoms, and coping strategies for ADRs caused by anticancer treatments will undoubtedly benefit more children.
Collapse
Affiliation(s)
- Hui Yan
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Penggao Wang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Fang Yang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Weyland Cheng
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Congcong Chen
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Bo Zhai
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yang Zhou
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| |
Collapse
|
26
|
Vahidian F, Lamaze FC, Bouffard C, Coulombe F, Gagné A, Blais F, Tonneau M, Orain M, Routy B, Manem VSK, Joubert P. CXCL13 Positive Cells Localization Predict Response to Anti-PD-1/PD-L1 in Pulmonary Non-Small Cell Carcinoma. Cancers (Basel) 2024; 16:708. [PMID: 38398098 PMCID: PMC10887067 DOI: 10.3390/cancers16040708] [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: 11/18/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) have revolutionized non-small cell lung cancers (NSCLCs) treatment, but only 20-30% of patients benefit from these treatments. Currently, PD-L1 expression in tumor cells is the only clinically approved predictor of ICI response in lung cancer, but concerns arise due to its low negative and positive predictive value. Recent studies suggest that CXCL13+ T cells in the tumor microenvironment (TME) may be a good predictor of response. We aimed to assess if CXCL13+ cell localization within the TME can predict ICI response in advanced NSCLC patients. Methods: This retrospective study included 65 advanced NSCLC patients treated with Nivolumab/Pembrolizumab at IUCPQ or CHUM and for whom a pretreatment surgical specimen was available. Good responders were defined as having a complete radiologic response at 1 year, and bad responders were defined as showing cancer progression at 1 year. IHC staining for CXCL13 was carried out on a representative slide from a resection specimen, and CXCL13+ cell density was evaluated in tumor (T), invasive margin (IM), non-tumor (NT), and tertiary lymphoid structure (TLS) compartments. Cox models were used to analyze progression-free survival (PFS) and overall survival (OS) probability, while the Mann-Whitney test was used to compare CXCL13+ cell density between responders and non-responders. Results: We showed that CXCL13+ cell density localization within the TME is associated with ICI efficacy. An increased density of CXCL13+ cells across all compartments was associated with a poorer prognostic (OS; HR = 1.22; 95%CI = 1.04-1.42; p = 0.01, PFS; HR = 1.16; p = 0.02), or a better prognostic when colocalized within TLSs (PFS; HR = 0.84, p = 0.03). Conclusion: Our results support the role of CXCL13+ cells in advanced NSCLC patients, with favorable prognosis when localized within TLSs and unfavorable prognosis when present elsewhere. The concomitant proximity of CXCL13+ and CD20+ cells within TLSs may favor antigen presentation to T cells, thus enhancing the effect of PD-1/PD-L1 axis inhibition. Further validation is warranted to confirm the potential relevance of this biomarker in a clinical setting.
Collapse
Affiliation(s)
- Fatemeh Vahidian
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Fabien C. Lamaze
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
| | - Cédrik Bouffard
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - François Coulombe
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Andréanne Gagné
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Florence Blais
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| | - Marion Tonneau
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.T.)
| | - Michèle Orain
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.T.)
| | - Venkata S. K. Manem
- Centre de Recherche du CHU de Québec—Université Laval, Quebec City, QC G1V 4G5, Canada
- Department of Mathematics and Computer Science, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Philippe Joubert
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec City, QC G1V 4G5, Canada (F.C.L.); (M.O.)
- Faculty of Medicine, Laval University, Quebec City, QC G1V 4G5, Canada (F.B.)
| |
Collapse
|
27
|
Zhou J, Du H, Cai W. Narrative review: precision medicine applications in neuroblastoma-current status and future prospects. Transl Pediatr 2024; 13:164-177. [PMID: 38323175 PMCID: PMC10839273 DOI: 10.21037/tp-23-557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Background and Objective Neuroblastoma (NB) is a common malignant tumor in children, and its treatment remains challenging. Precision medicine, as an individualized treatment strategy, aims to improve efficacy and reduce toxicity by combining unique patient- and tumor-related factors, bringing new hope for NB treatment. In this article, we review the evidence related to precision medicine in NB, with a focus on potential clinically actionable targets and a series of targeted drugs associated with NB. Methods We conducted an extensive search in PubMed, EMBASE, and Web of Science using key terms and database-specific strategies, filtered for time and language, to ensure a comprehensive collection of literature related to precision medicine in NB. The main search terms consisted of "neuroblastoma", "precision medicine", "pediatrics", and "targeting". The articles included in this study encompass those published from 1985 to the present, without restrictions on the type of articles. Key Content and Findings ALK inhibitors and MYCN inhibitors have been developed to interfere with tumor cell growth and dissemination, thereby improving treatment outcomes. Additionally, systematic testing to identify relevant driver mutations is crucial and can be used for diagnosis and prognostic assessment through the detection of many associated molecular markers. Furthermore, liquid biopsy, a non-invasive tumor detection method, can complement tissue biopsy and play a role in NB by analyzing circulating tumor DNA and circulating tumor cells to provide genetic information and molecular characteristics of the tumor. Recently, trials conducted by many pediatric oncology groups have shown the urgent need for new approaches to cure relapsed and refractory patients. Conclusions The purpose of this review is to summarize the latest advances in clinical treatment of NB, to better understand and focus on the development of promising treatment approaches, and to expedite the transition to the precision medicine clinical relevance in NB patients.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongmei Du
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weisong Cai
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
28
|
Patel AG, Ashenberg O, Collins NB, Segerstolpe Å, Jiang S, Slyper M, Huang X, Caraccio C, Jin H, Sheppard H, Xu K, Chang TC, Orr BA, Shirinifard A, Chapple RH, Shen A, Clay MR, Tatevossian RG, Reilly C, Patel J, Lupo M, Cline C, Dionne D, Porter CBM, Waldman J, Bai Y, Zhu B, Barrera I, Murray E, Vigneau S, Napolitano S, Wakiro I, Wu J, Grimaldi G, Dellostritto L, Helvie K, Rotem A, Lako A, Cullen N, Pfaff KL, Karlström Å, Jané-Valbuena J, Todres E, Thorner A, Geeleher P, Rodig SJ, Zhou X, Stewart E, Johnson BE, Wu G, Chen F, Yu J, Goltsev Y, Nolan GP, Rozenblatt-Rosen O, Regev A, Dyer MA. A spatial cell atlas of neuroblastoma reveals developmental, epigenetic and spatial axis of tumor heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574538. [PMID: 38260392 PMCID: PMC10802404 DOI: 10.1101/2024.01.07.574538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuroblastoma is a pediatric cancer arising from the developing sympathoadrenal lineage with complex inter- and intra-tumoral heterogeneity. To chart this complexity, we generated a comprehensive cell atlas of 55 neuroblastoma patient tumors, collected from two pediatric cancer institutions, spanning a range of clinical, genetic, and histologic features. Our atlas combines single-cell/nucleus RNA-seq (sc/scRNA-seq), bulk RNA-seq, whole exome sequencing, DNA methylation profiling, spatial transcriptomics, and two spatial proteomic methods. Sc/snRNA-seq revealed three malignant cell states with features of sympathoadrenal lineage development. All of the neuroblastomas had malignant cells that resembled sympathoblasts and the more differentiated adrenergic cells. A subset of tumors had malignant cells in a mesenchymal cell state with molecular features of Schwann cell precursors. DNA methylation profiles defined four groupings of patients, which differ in the degree of malignant cell heterogeneity and clinical outcomes. Using spatial proteomics, we found that neuroblastomas are spatially compartmentalized, with malignant tumor cells sequestered away from immune cells. Finally, we identify spatially restricted signaling patterns in immune cells from spatial transcriptomics. To facilitate the visualization and analysis of our atlas as a resource for further research in neuroblastoma, single cell, and spatial-omics, all data are shared through the Human Tumor Atlas Network Data Commons at www.humantumoratlas.org.
Collapse
Affiliation(s)
- Anand G Patel
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- These authors contributed equally
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- These authors contributed equally
| | - Natalie B Collins
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| | - Åsa Segerstolpe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sizun Jiang
- Department of Pathology, Stanford University, Stanford, CA, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Huang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chiara Caraccio
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heather Sheppard
- Comparative Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ke Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard H Chapple
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amber Shen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael R Clay
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ruth G Tatevossian
- Cancer Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Colleen Reilly
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jaimin Patel
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marybeth Lupo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Cynthia Cline
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Caroline B M Porter
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yunhao Bai
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Bokai Zhu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Evan Murray
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sébastien Vigneau
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara Napolitano
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Isaac Wakiro
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jingyi Wu
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Grace Grimaldi
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laura Dellostritto
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karla Helvie
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Asaf Rotem
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ana Lako
- Center for Immuno-Oncology (CIO), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicole Cullen
- Center for Immuno-Oncology (CIO), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Center for Immuno-Oncology (CIO), Dana-Farber Cancer Institute, Boston, MA, USA
| | - Åsa Karlström
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Judit Jané-Valbuena
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ellen Todres
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron Thorner
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul Geeleher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Elizabeth Stewart
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bruce E Johnson
- Center for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Fei Chen
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yury Goltsev
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Current address: Research and Early Development, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Current address: Research and Early Development, Genentech Inc., South San Francisco, CA, 94080, USA
- Lead contacts
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Lead contacts
| |
Collapse
|
29
|
Moreno L, Weston R, Owens C, Valteau-Couanet D, Gambart M, Castel V, Zwaan CM, Nysom K, Gerber N, Castellano A, Laureys G, Ladenstein R, Rössler J, Makin G, Murphy D, Morland B, Vaidya S, Thebaud E, van Eijkelenburg N, Tweddle DA, Barone G, Tandonnet J, Corradini N, Chastagner P, Paillard C, Bautista FJ, Gallego Melcon S, De Wilde B, Marshall L, Gray J, Burchill SA, Schleiermacher G, Chesler L, Peet A, Leach MO, McHugh K, Hayes R, Jerome N, Caron H, Laidler J, Fenwick N, Holt G, Moroz V, Kearns P, Gates S, Pearson ADJ, Wheatley K. Bevacizumab, Irinotecan, or Topotecan Added to Temozolomide for Children With Relapsed and Refractory Neuroblastoma: Results of the ITCC-SIOPEN BEACON-Neuroblastoma Trial. J Clin Oncol 2024:JCO2300458. [PMID: 38190578 DOI: 10.1200/jco.23.00458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/25/2023] [Accepted: 10/05/2023] [Indexed: 01/10/2024] Open
Abstract
PURPOSE Outcomes for children with relapsed and refractory high-risk neuroblastoma (RR-HRNB) remain dismal. The BEACON Neuroblastoma trial (EudraCT 2012-000072-42) evaluated three backbone chemotherapy regimens and the addition of the antiangiogenic agent bevacizumab (B). MATERIALS AND METHODS Patients age 1-21 years with RR-HRNB with adequate organ function and performance status were randomly assigned in a 3 × 2 factorial design to temozolomide (T), irinotecan-temozolomide (IT), or topotecan-temozolomide (TTo) with or without B. The primary end point was best overall response (complete or partial) rate (ORR) during the first six courses, by RECIST or International Neuroblastoma Response Criteria for patients with measurable or evaluable disease, respectively. Safety, progression-free survival (PFS), and overall survival (OS) time were secondary end points. RESULTS One hundred sixty patients with RR-HRNB were included. For B random assignment (n = 160), the ORR was 26% (95% CI, 17 to 37) with B and 18% (95% CI, 10 to 28) without B (risk ratio [RR], 1.52 [95% CI, 0.83 to 2.77]; P = .17). Adjusted hazard ratio for PFS and OS were 0.89 (95% CI, 0.63 to 1.27) and 1.01 (95% CI, 0.70 to 1.45), respectively. For irinotecan ([I]; n = 121) and topotecan (n = 60) random assignments, RRs for ORR were 0.94 and 1.22, respectively. A potential interaction between I and B was identified. For patients in the bevacizumab-irinotecan-temozolomide (BIT) arm, the ORR was 23% (95% CI, 10 to 42), and the 1-year PFS estimate was 0.67 (95% CI, 0.47 to 0.80). CONCLUSION The addition of B met protocol-defined success criteria for ORR and appeared to improve PFS. Within this phase II trial, BIT showed signals of antitumor activity with acceptable tolerability. Future trials will confirm these results in the chemoimmunotherapy era.
Collapse
Affiliation(s)
- Lucas Moreno
- Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | - Guy Makin
- Central Manchester and Manchester Children's University Hospitals NHS Trust, Manchester, United Kingdom
| | - Dermot Murphy
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Bruce Morland
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Sucheta Vaidya
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | | | | | - Deborah A Tweddle
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | | | | | | | | | | | | | | | | | - Lynley Marshall
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | - Juliet Gray
- University Hospital Southampton, Southampton, United Kingdom
| | | | | | - Louis Chesler
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | - Andrew Peet
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Martin O Leach
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | - Kieran McHugh
- Great Ormond Street Hospital, London, United Kingdom
| | | | - Neil Jerome
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | | | | | | | - Grace Holt
- University of Birmingham, Birmingham, United Kingdom
| | | | - Pamela Kearns
- University of Birmingham, Birmingham, United Kingdom
| | - Simon Gates
- University of Birmingham, Birmingham, United Kingdom
| | - Andrew D J Pearson
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | | |
Collapse
|
30
|
Hui Z, Wen H, Zhu J, Deng H, Jiang X, Ye XY, Wang L, Xie T, Bai R. Discovery of plant-derived anti-tumor natural products: Potential leads for anti-tumor drug discovery. Bioorg Chem 2024; 142:106957. [PMID: 37939507 DOI: 10.1016/j.bioorg.2023.106957] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/14/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023]
Abstract
Natural products represent a paramount source of novel drugs. Numerous plant-derived natural products have demonstrated potent anti-tumor properties, thereby garnering considerable interest in their potential as anti-tumor drugs. This review compiles an overview of 242 recently discovered natural products, spanning the period from 2018 to the present. These natural products, which include 69 terpenoids, 42 alkaloids, 39 flavonoids, 21 steroids, 14 phenylpropanoids, 5 quinolines and 52 other compounds, are characterized by their respective chemical structures, anti-tumor activities, and mechanisms of action. By providing an essential reference and fresh insights, this review aims to support and inspire researchers engaged in the fields of natural products and anti-tumor drug discovery.
Collapse
Affiliation(s)
- Zi Hui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Hao Wen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Junlong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Haowen Deng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Liwei Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China.
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China.
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China.
| |
Collapse
|
31
|
Kushner BH, LaQuaglia MP, Cardenas FI, Basu EM, Gerstle JT, Kramer K, Roberts SS, Wolden SL, Cheung NKV, Modak S. Stage 4N neuroblastoma before and during the era of anti-G D2 immunotherapy. Int J Cancer 2023; 153:2019-2031. [PMID: 37602920 DOI: 10.1002/ijc.34693] [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: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023]
Abstract
Patients with stage 4N neuroblastoma (distant metastases limited to lymph nodes) stand out as virtually the only survivors of high-risk neuroblastoma (HR-NB) before myeloablative therapy (MAT) and immunotherapy with anti-GD2 monoclonal antibodies (mAbs) became standard. Because no report presents more recent results with 4N, we analyzed our large 4N experience. All 51 pediatric 4N patients (<18 years old) diagnosed 1985 to 2021 were reviewed. HR-NB included MYCN-nonamplified 4N diagnosed at age ≥18 months and MYCN-amplified 4N. Among 34 MYCN-nonamplified high-risk patients, 20 are relapse-free 1.5+ to 37.5+ (median 12.5+) years post-diagnosis, including 13 without prior MAT and 5 treated with little (1 cycle; n = 2) or no mAb (n = 3), while 14 patients (7 post-MAT, 8 post-mAbs) relapsed (all soft tissue). Of 15 MYCN-amplified 4N patients, 7 are relapse-free 2.1+ to 26.4+ (median 11.6+) years from the start of chemotherapy (all received mAbs; 3 underwent MAT) and 4 are in second remission 4.2+ to 21.8+ years postrelapse (all soft tissue). Statistical analyses showed no significant association of survival with either MAT or mAbs for MYCN-nonamplified HR-NB; small numbers prevented these analyses for MYCN-amplified patients. The two patients with intermediate-risk 4N (14-months-old) are relapse-free 7+ years postresection of primary tumors; distant disease spontaneously regressed. The natural history of 4N is marked by NB confined to soft tissue without early relapse in bones or bone marrow, where mAbs have proven efficacy. These findings plus curability without MAT, as seen elsewhere and at our center, support consideration of treatment reduction for MYCN-nonamplified 4N.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael P LaQuaglia
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Justin T Gerstle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
32
|
Shen AL, Zhao J, Yu LT, Zhang AA, Wu B, Fang Y, Han YL, Li CSZ, Li ZL, Gao YJ, Zhang SG. Cost-effectiveness analysis of dinutuximab β for the treatment of high-risk neuroblastoma in China. Pediatr Blood Cancer 2023; 70:e30680. [PMID: 37715719 DOI: 10.1002/pbc.30680] [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: 06/30/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Dinutuximab β can be used to treat children with high-risk neuroblastoma (NB). Due to its high price, whether dinutuximab β is cost-effective for the treatment of high-risk NB remains uncertain. Therefore, assessing the cost-effectiveness of dinutuximab β in children with high-risk NB is of high importance. METHODS The health utilities and economic outcomes in children with high-risk NB were projected using a partitioned survival model. The individual patient data (IPD) of add-on treatment with dinutuximab β (GD2 group) were derived from the literature, while the IPD of traditional therapy (TT group) were obtained from retrospective data of Shanghai Children's Medical Center. Treatment costs included drugs, adverse event-related expenses, and medical resource use. Utility values were obtained from the literature. Costs and quality-adjusted life-years (QALYs) were measured over a 10-year time horizon. Deterministic sensitivity analyses (DSA) and probabilistic sensitivity analyses (PSA) were also conducted. RESULTS Compared with the TT group, QALY increased in the GD2 group by 0.72 with an increased cost of $171,269.70, leading to an incremental cost-effectiveness ratio of 236,462.75$/QALY. DSA showed that the price of dinutuximab β was the main factor on the results than other parameters. Compared with the TT group, the GD2 group could not be cost-effective in the PSA at the $37,920/QALY threshold. CONCLUSION Results found that dinutuximab β is not a cost-effective treatment option for children with high-risk NB unless its price is significantly reduced.
Collapse
Affiliation(s)
- An-Le Shen
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhao
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Ting Yu
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - An-An Zhang
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Wu
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Fang
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Ya-Li Han
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Sui-Zi Li
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Ling Li
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Jin Gao
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shun-Guo Zhang
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
De Ioris MA, Fabozzi F, Del Bufalo F, Del Baldo G, Villani MF, Cefalo MG, Garganese MC, Stracuzzi A, Tangari F, Greco AM, Giovannoni I, Carta R, D'Andrea ML, Mastronuzzi A, Locatelli F. Venetoclax plus cyclophosphamide and topotecan in heavily pre-treated relapsed metastatic neuroblastoma: a single center case series. Sci Rep 2023; 13:19295. [PMID: 37935707 PMCID: PMC10630499 DOI: 10.1038/s41598-023-44993-9] [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: 08/03/2023] [Accepted: 10/14/2023] [Indexed: 11/09/2023] Open
Abstract
The prognosis of relapsed/refractory (R/R) neuroblastoma (NB) is dismal, calling for new therapeutic strategies. Venetoclax (VEN) is a highly selective, potent, orally bioavailable, BCL-2 inhibitor small-molecule that showed a synergistic effect with cyclophosphamide and topotecan (Cy-Topo) in murine NB models. Our aim was to evaluate the feasibility of VEN plus Cy-Topo in children with R/R NB. Four patients, who had previously failed > 3 lines of treatment, were treated with VEN plus Cy-Topo based on a 28-day schedule in an outpatient setting. BCL-2 expression in immunochemistry on tumor samples at relapse and the BCL2 gene status was evaluated in all patients. The main toxicity was hematological, with grade 4 neutropenia and thrombocytopenia occurring in all courses and leading to transient VEN discontinuation. Grade 3 oral mucositis was observed in 1/8 courses. No other grade 2-4 toxicities were observed. BCL-2 was expressed in all tumors, while no molecular abnormalities in the BCL-2 genes were detected. A stable disease was observed in all patients, without any progression during the study period. VEN plus Cy-Topo is well tolerated, with encouraging results that may be improved by testing the schedule in less advanced patients.
Collapse
Affiliation(s)
- Maria Antonietta De Ioris
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Francesco Fabozzi
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Del Bufalo
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giada Del Baldo
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Maria Giuseppina Cefalo
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | | - Federica Tangari
- Unit of Clinical Pharmacy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Arturo Maria Greco
- Unit of Clinical Pharmacy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Roberto Carta
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Angela Mastronuzzi
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology and of Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Milan, Italy
| |
Collapse
|
34
|
Lode HN, Ehlert K, Huber S, Troschke-Meurer S, Siebert N, Zumpe M, Loibner H, Ladenstein R. Long-term, continuous infusion of single-agent dinutuximab beta for relapsed/refractory neuroblastoma: an open-label, single-arm, Phase 2 study. Br J Cancer 2023; 129:1780-1786. [PMID: 37813959 PMCID: PMC10667538 DOI: 10.1038/s41416-023-02457-x] [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: 05/05/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Short-term infusions of dinutuximab beta plus isotretinoin and cytokines administered in previous immunotherapy studies in neuroblastoma were associated with severe pain. Here, long-term, continuous infusion of single-agent dinutuximab beta was evaluated in patients with relapsed/refractory neuroblastoma. METHODS In this open-label, single-arm, Phase 2 study, patients with either refractory or relapsed high-risk neuroblastoma received dinutuximab beta by continuous infusion over 10 days of each cycle, for up to five cycles. The primary endpoint was objective response rate 24 weeks after the end of cycle 5. Secondary endpoints included adverse events, intravenous morphine use, best response, duration of response, and three-year progression-free and overall survival. RESULTS Of the 40 patients included, 38 had evaluable response. Objective response rate was 26% and best response rate 37%. Median duration of response was 238 days (IQR 108-290). Three-year progression-free and overall survival rates were 31% (95% CI 17-47) and 66% (95% CI 47-79), respectively. Prophylactic intravenous morphine use and duration of use decreased with increasing cycles. The most common grade 3 treatment-related adverse events were pain, diarrhea, and hypokalemia. CONCLUSION Long-term continuous infusion of single-agent dinutuximab beta is tolerable and associated with clinically meaningful responses in patients with relapsed/refractory high-risk neuroblastoma. CLINICAL TRIAL REGISTRATION The study is registered with ClinicalTrials.gov (NCT02743429) and EudraCT (2014-000588-42).
Collapse
Grants
- This research was funded by the University Medicine Greifswald, H.W. & J. Hector Stiftungen, Germany, under Grant M2116, Apeiron Biologics, Vienna, Austria under Grant APN, and Apeiron (Vienna, Austria) providing dinutuximab beta (ch14.18/CHO), and the St. Anna Kinderkrebsforschung (Vienna, Austria). Further funding was provided by EUSA Pharma (Hemel Hempstead, UK), which has marketing authorization for dinutuximab beta in Europe. Editorial assistance for the development of the manuscript was funded by EUSA Pharma
Collapse
Affiliation(s)
- Holger N Lode
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany.
| | - Karoline Ehlert
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Stephanie Huber
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Sascha Troschke-Meurer
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Nikolai Siebert
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Maxi Zumpe
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | | | - Ruth Ladenstein
- Department of Studies and Statistics for Integrated Research and Projects, Children's Cancer Research Institute, Vienna, Austria
- Department of Paediatrics, St Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Sevrin F, Kolesnikov-Gauthier H, Cougnenc O, Bogart E, Schleiermacher G, Courbon F, Gambart M, Giraudet AL, Corradini N, Badel JN, Rault E, Oudoux A, Deley MCL, Valteau-Couanet D, Defachelles AS. Phase II study of 131 I-metaiodobenzylguanidine with 5 days of topotecan for refractory or relapsed neuroblastoma: Results of the French study MIITOP. Pediatr Blood Cancer 2023; 70:e30615. [PMID: 37574821 DOI: 10.1002/pbc.30615] [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: 05/03/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023]
Abstract
PURPOSE We report the results of the French multicentric phase II study MIITOP (NCT00960739), which evaluated tandem infusions of 131 I-metaiodobenzylguanidine (mIBG) and topotecan in children with relapsed/refractory metastatic neuroblastoma (NBL). METHODS Patients received 131 I-mIBG on day 1, with intravenous topotecan daily on days 1-5. A second activity of 131 I-mIBG was given on day 21 to deliver a whole-body radiation dose of 4 Gy, combined with a second course of topotecan on days 21-25. Peripheral blood stem cells were infused on day 31. RESULTS Thirty patients were enrolled from November 2008 to June 2015. Median age at diagnosis was 5.5 years (2-20). Twenty-one had very high-risk NBL (VHR-NBL), that is, stage 4 NBL at diagnosis or at relapse, with insufficient response (i.e., less than a partial response of metastases and more than three mIBG spots) after induction chemotherapy; nine had progressive metastatic relapse. Median Curie score at inclusion was 6 (1-26). Median number of prior lines of treatment was 3 (1-7). Objective response rate was 13% (95% confidence interval [CI]: 4-31) for the whole population, 19% for VHR-NBL, and 0% for progressive relapses. Immediate tolerance was good, with nonhematologic toxicity limited to grade-2 nausea/vomiting in eight patients. Two-year event-free survival was 17% (95% CI: 6-32). Among the 16 patients with VHR-NBL who had not received prior myeloablative busulfan-melphalan consolidation, 13 had at least stable disease after MIITOP; 11 subsequently received busulfan-melphalan; four of them were alive (median follow-up: 7 years). CONCLUSION MIITOP showed acceptable tolerability in this heavily pretreated population and encouraging survival rates in VHR-NBL when followed by busulfan-melphalan.
Collapse
Affiliation(s)
- François Sevrin
- Department of Pediatric Oncology, Oscar Lambret Center, Lille, France
| | | | - Olivier Cougnenc
- Department of Clinical Pharmacy, Oscar Lambret Center, Lille, France
| | - Emilie Bogart
- Department of Methodology and Biostatistics, Oscar Lambret Center, Lille, France
| | | | - Frederic Courbon
- Service de Médecine Nucléaire, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Marion Gambart
- Hematology and Oncology Unit, Children's Hospital, CHU Toulouse, Toulouse, France
| | | | - Nadège Corradini
- Institute of Pediatric Hematology and Oncology, Léon Bérard Center, Lyon, France
| | - Jean-Noël Badel
- Department of Nuclear Medicine, Léon Bérard Center, Lyon, France
| | - Erwann Rault
- Department of Medical Physics, Oscar Lambret Center, Lille, France
| | - Aurore Oudoux
- Department of Nuclear Medicine, Oscar Lambret Center, Lille, France
| | | | | | | |
Collapse
|
36
|
Jacob M, Wiedemann S, Brücher D, Pieper NM, Birkhold M, Särchen V, Jeroch J, Demes MC, Gretser S, Braun Y, Gradhand E, Rothweiler F, Michaelis M, Cinatl J, Vogler M. Increased MCL1 dependency leads to new applications of BH3-mimetics in drug-resistant neuroblastoma. Br J Cancer 2023; 129:1667-1678. [PMID: 37723317 PMCID: PMC10646009 DOI: 10.1038/s41416-023-02430-8] [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: 03/14/2023] [Revised: 08/16/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Neuroblastoma is a paediatric cancer that is characterised by poor prognosis for chemoresistant disease, highlighting the need for better treatment options. Here, we asked whether BH3-mimetics inhibiting BCL2 proteins may eliminate chemoresistant neuroblastoma cells. METHODS We utilised cisplatin-adapted neuroblastoma cell lines as well as patient tissues before and after relapse to study alterations of BCL2 proteins upon chemoresistance. RESULTS In a direct comparison of cisplatin-resistant cells we identified a prominent loss of sensitivity to BCL2/BCL-XL inhibitors that is associated with an increase in MCL1 dependency and high expression of MCL1 in patient tumour tissues. Screening of FDA-approved anti-cancer drugs in chemoresistant cells identified therapeutics that may be beneficial in combination with the clinically tested BH3-mimetic ABT263, but no synergistic drug interactions with the selective MCL1 inhibitor S63845. Further exploration of potential treatment options for chemoresistant neuroblastoma identified immunotherapy based on NK cells as highly promising, since NK cells are able to efficiently kill both parental and chemoresistant cells. CONCLUSIONS These data highlight that the application of BH3-mimetics may differ between first line treatment and relapsed disease. Combination of NK cell-based immunotherapy with BH3-mimetics may further increase killing of chemoresistant neuroblastoma, outlining a new treatment strategy for relapsed neuroblastoma.
Collapse
Affiliation(s)
- Maureen Jacob
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Sara Wiedemann
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Daniela Brücher
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Nadja M Pieper
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Moni Birkhold
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Vinzenz Särchen
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Jan Jeroch
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Melanie C Demes
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Steffen Gretser
- Department of Pediatric and Perinatal Pathology, Dr. Senckenberg Institute of Pathology, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Yannick Braun
- Department of Pediatric Surgery and Pediatric Urology, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Elise Gradhand
- Department of Pediatric and Perinatal Pathology, Dr. Senckenberg Institute of Pathology, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Florian Rothweiler
- Institute for Medical Virology, Goethe-University Frankfurt, Frankfurt am Main, Germany
- Dr. Petra Joh-Forschungshaus, Frankfurt am Main, Germany
| | - Martin Michaelis
- Dr. Petra Joh-Forschungshaus, Frankfurt am Main, Germany
- School of Biosciences, University of Kent, Canterbury, UK
| | - Jindrich Cinatl
- Institute for Medical Virology, Goethe-University Frankfurt, Frankfurt am Main, Germany
- Dr. Petra Joh-Forschungshaus, Frankfurt am Main, Germany
| | - Meike Vogler
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.
| |
Collapse
|
37
|
Muñoz JP, Larrosa C, Chamorro S, Perez-Jaume S, Simao M, Sanchez-Sierra N, Varo A, Gorostegui M, Castañeda A, Garraus M, Lopez-Miralles S, Mora J. Early Salvage Chemo-Immunotherapy with Irinotecan, Temozolomide and Naxitamab Plus GM-CSF (HITS) for Patients with Primary Refractory High-Risk Neuroblastoma Provide the Best Chance for Long-Term Outcomes. Cancers (Basel) 2023; 15:4837. [PMID: 37835531 PMCID: PMC10571514 DOI: 10.3390/cancers15194837] [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] [Received: 08/24/2023] [Revised: 09/26/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023] Open
Abstract
Patients with high-risk neuroblastoma (HR-NB) who are unable to achieve a complete response (CR) to induction therapy have worse outcomes. We investigated the combination of humanized anti-GD2 mAb naxitamab (Hu3F8), irinotecan (I), temozolomide (T), and sargramostim (GM-CSF)-HITS-against primary resistant HR-NB. Eligibility criteria included having a measurable chemo-resistant disease at the end of induction (EOI) treatment. Patients were excluded if they had progressive disease (PD) during induction. Prior anti-GD2 mAb and/or I/T therapy was permitted. Each cycle, administered four weeks apart, comprised Irinotecan 50 mg/m2/day intravenously (IV) plus Temozolomide 150 mg/m2/day orally (days 1-5); naxitamab 2.25 mg/kg/day IV on days 2, 4, 8 and 10, (total 9 mg/kg or 270 mg/m2 per cycle), and GM-CSF 250 mg/m2/day subcutaneously was used (days 6-10). Toxicity was measured using CTCAE v4.0 and responses through the modified International Neuroblastoma Response Criteria (INRC). Thirty-four patients (median age at treatment initiation, 4.9 years) received 164 (median 4; 1-12) HITS cycles. Toxicities included myelosuppression and diarrhea, which was expected with I/T, and pain and hypertension, expected with naxitamab. Grade ≥3-related toxicities occurred in 29 (85%) of the 34 patients; treatment was outpatient. The best responses were CR = 29% (n = 10); PR = 3% (n = 1); SD = 53% (n = 18); PD = 5% (n = 5). For cohort 1 (early treatment), the best responses were CR = 47% (n = 8) and SD = 53% (n = 9). In cohort 2 (late treatment), the best responses were CR = 12% (n = 2); PR = 6% (n = 1); SD = 53% (n = 9); and PD = 29% (n = 5). Cohort 1 had a 3-year OS of 84.8% and EFS 54.4%, which are statistically significant improvements (EFS p = 0.0041 and OS p = 0.0037) compared to cohort 2. In conclusion, naxitamab-based chemo-immunotherapy is effective against primary chemo-resistant HR-NB, increasing long-term outcomes when administered early during the course of treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (J.P.M.); (C.L.); (S.C.); (S.P.-J.); (M.S.); (N.S.-S.); (A.V.); (M.G.); (A.C.); (M.G.); (S.L.-M.)
| |
Collapse
|
38
|
Mohd AB, Mohd OB, Alabdallat YJ, Al Dwairy SY, Ghannam RA, Hanaqtah BM, Albakri KA. Safety and efficacy of dinutuximab in the treatment of neuroblastoma: A review. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2023; 28:71. [PMID: 38116487 PMCID: PMC10729685 DOI: 10.4103/jrms.jrms_727_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 07/11/2023] [Accepted: 08/07/2023] [Indexed: 12/21/2023]
Abstract
Dinutuximab, which is a monoclonal antibody targeting GD2 expressed in neuroblasts, improves survival when included in the therapy regimen. This article reviews the importance of dinutuximab in managing neuroblastoma (NB). Dinutuximab targets high levels of GD2 expression in NB cells, thus increasing event-free survival when used in the maintenance therapy of high-risk patients with NB. Although several collaborative studies have set the standard of care for maintenance therapy, the long-term follow-up and continuous evaluation of the use of antibodies and the co-administration of other pharmacological or immunomodulatory drugs remain to be studied. Trials have shown that the use of dinutuximab for maintenance therapy can prolong the time before the first relapse and improve overall survival. However, there is uncertainty in the function of cytokines co-administered with dinutuximab, which may lead to increased toxicity without additional benefits. Recent studies on relapsed and refractory NB have shown the potential efficacy of dinutuximab. Further research is required to properly incorporate Dinutuximab in current treatment modalities.
Collapse
Affiliation(s)
- Ahmed Bassam Mohd
- Medicine, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
| | - Omar B Mohd
- Medicine, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
| | - Yasmeen J Alabdallat
- Medicine, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
| | - Salem Yousef Al Dwairy
- Department of Special Surgery, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
- Neurosurgery Division, Department of Surgery, Prince Hamzah Hospital, Prince Hamzah Street, Al-Rewaq, Amman, 11732, Jordan
| | - Reem A Ghannam
- Medicine, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
| | - Balqees M Hanaqtah
- Medicine, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
| | - Khaled A Albakri
- Medicine, Faculty of Medicine, The Hashemite University, P.O.Box 330127, Zarqa, 13133, Jordan
| |
Collapse
|
39
|
Wahba A, Wolters R, Foster JH. Neuroblastoma in the Era of Precision Medicine: A Clinical Review. Cancers (Basel) 2023; 15:4722. [PMID: 37835416 PMCID: PMC10571527 DOI: 10.3390/cancers15194722] [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: 06/28/2023] [Revised: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 10/15/2023] Open
Abstract
The latest advances in treatment for patients with neuroblastoma are constantly being incorporated into clinical trials and clinical practice standards, resulting in incremental improvements in the survival of patients over time. Survivors of high-risk neuroblastoma (HRNBL), however, continue to develop treatment-related late effects. Additionally, for the majority of the nearly 50% of patients with HRNBL who experience relapse, no curative therapy currently exists. As technologies in diagnostic and molecular profiling techniques rapidly advance, so does the discovery of potential treatment targets. Here, we discuss the current clinical landscape of therapies for neuroblastoma in the era of precision medicine.
Collapse
Affiliation(s)
| | | | - Jennifer H. Foster
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Cancer Center, Houston, TX 77030, USA; (A.W.); (R.W.)
| |
Collapse
|
40
|
Cupit-Link M, Federico SM. Treatment of High-Risk Neuroblastoma with Dinutuximab and Chemotherapy Administered in all Cycles of Induction. Cancers (Basel) 2023; 15:4609. [PMID: 37760578 PMCID: PMC10527563 DOI: 10.3390/cancers15184609] [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: 08/09/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Administration of chemoimmunotherapy using concurrent chemotherapy and an anti-GD2 monoclonal antibody (mAb), dinutuximab (DIN), demonstrated efficacy for the treatment of relapsed and refractory neuroblastoma. Chemoimmunotherapy, using a humanized anti-GD2 mAb, demonstrated a signal of activity in a phase 2 study for the treatment of patients with newly diagnosed high-risk neuroblastoma (HRNBL). In this single-institution retrospective study, patients with HRNBL received an Induction chemotherapy regimen plus DIN in all Induction cycles. Toxicity and response data were abstracted from the electronic medical record. Toxicities were graded by CTCAE v.5.0. The end of Induction (EOI) objective response rate was determined using the Revised International Neuroblastoma Response Criteria. Twenty-seven patients with HRNBL (23 newly diagnosed, 16 females, median age 3.9 years) started Induction chemoimmunotherapy from 27 January 2017 to 28 December 2022. All patients received DIN with all cycles of Induction therapy, and all but one patient completed Induction therapy. The most common non-hematologic grade ≥ 3 toxicities included fever (44%), hypoxemia (20%), and hypoalbuminemia (11%). End of Induction responses included eighteen with a complete response (CR), seven with a partial response (PR), one with progressive disease (PD), and zero with a minor response or stable disease. Twenty-six of twenty-seven patients (96%) completed all Induction cycles and were evaluable for a response. The EOI response of PR or better in the evaluable cohort was 96%. Dinutuximab was well tolerated with all Induction cycles, demonstrated an encouraging EOI response rate, and should be evaluated in a randomized study.
Collapse
|
41
|
Cupit-Link M, Hagiwara K, Zhang J, Federico SM. Clinical Response to a PARP Inhibitor and Chemotherapy in a Child with BARD1-Mutated Refractory Neuroblastoma: A Case Report. RESEARCH SQUARE 2023:rs.3.rs-3250117. [PMID: 37645774 PMCID: PMC10462232 DOI: 10.21203/rs.3.rs-3250117/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Despite advances in the treatment of high-risk neuroblastoma, approximately half of these patients die from the disease. Targeted therapy based on synthetic lethality associated with homologous recombination deficiency (HRD) caused by germline mutations in homologous recombination repair genes has shown great efficacy in several adult solid tumors. Here we report the first successful treatment of a pediatric patient with refractory neuroblastoma and a germline pathogenic mutation in BARD1 using a PARP inhibitor, talazoparib, in combination with cytotoxic chemotherapy and radiation therapy. Allele-specific expression in RNA-seq indicates bi-allelic loss of BARD1 in tumor; however, the HRD score was below the threshold currently used for HRD classification in adult cancers. Our study demonstrates that the use of PARP inhibition in combination with DNA-damaging agents should be considered in children with BARD1-mutated neuroblastoma and cautions against the use of HRD score alone as a biomarker for this pediatric population.
Collapse
Affiliation(s)
- Maggie Cupit-Link
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Kohei Hagiwara
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Sara M. Federico
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
42
|
Pilgrim AA, Jonus HC, Ho A, Cole AC, Shim J, Goldsmith KC. The yes-associated protein (YAP) is associated with resistance to anti-GD2 immunotherapy in neuroblastoma through downregulation of ST8SIA1. Oncoimmunology 2023; 12:2240678. [PMID: 37554309 PMCID: PMC10405770 DOI: 10.1080/2162402x.2023.2240678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023] Open
Abstract
Pediatric patients with high-risk neuroblastoma often relapse with chemotherapy-resistant, incurable disease. Relapsed neuroblastomas harbor chemo-resistant mesenchymal tumor cells and increased expression/activity of the transcriptional co-regulator, the Yes-Associated Protein (YAP). Patients with relapsed neuroblastoma are often treated with immunotherapy such as the anti-GD2 antibody, dinutuximab, in combination with chemotherapy. We have previously shown that YAP mediates both chemotherapy and MEK inhibitor resistance in relapsed RAS mutated neuroblastoma and so posited that YAP might also be involved in anti-GD2 antibody resistance. We now show that YAP genetic inhibition significantly enhances sensitivity of mesenchymal neuroblastomas to dinutuximab and gamma delta (γδ) T cells both in vitro and in vivo. Mechanistically, YAP inhibition induces increased GD2 cell surface expression through upregulation of ST8SIA1, the gene encoding GD3 synthase and the rate-limiting enzyme in GD2 biosynthesis. The mechanism of ST8SIA1 suppression by YAP is independent of PRRX1 expression, a mesenchymal master transcription factor, suggesting YAP may be the downstream effector of mesenchymal GD2 resistance. These results therefore identify YAP as a therapeutic target to augment GD2 immunotherapy responses in patients with neuroblastoma.
Collapse
Affiliation(s)
- Adeiye A. Pilgrim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Hunter C. Jonus
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew Ho
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna C. Cole
- Division of Surgical Oncology, Department of Surgery, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jenny Shim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, The Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - Kelly C. Goldsmith
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, The Children’s Healthcare of Atlanta, Atlanta, Georgia
| |
Collapse
|
43
|
Krystal J, Foster JH. Treatment of High-Risk Neuroblastoma. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1302. [PMID: 37628301 PMCID: PMC10453838 DOI: 10.3390/children10081302] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
High-risk neuroblastoma is a highly aggressive solid tumor that most commonly presents in early childhood. Advances in treatment through decades of clinical trials and research have led to improved outcomes. This review provides an overview of the current state of treatment for high-risk neuroblastoma.
Collapse
Affiliation(s)
- Julie Krystal
- Zucker Hofstra School of Medicine, Department of Pediatrics, Cohen Children’s Medical Center, New Hyde Park, NY 11040, USA
| | - Jennifer H. Foster
- Department of Pediatrics, Baylor College of Medicine, Texas Children’s Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|
44
|
Lode HN, Ladenstein R, Troschke-Meurer S, Struppe L, Siebert N, Zumpe M, Ehlert K, Huber S, Glogova E, Hundsdoerfer P, Eggert A, Zaniewska-Tekieli A, Balwierz W, Wieczorek A. Effect and Tolerance of N5 and N6 Chemotherapy Cycles in Combination with Dinutuximab Beta in Relapsed High-Risk Neuroblastoma Patients Who Failed at Least One Second-Line Therapy. Cancers (Basel) 2023; 15:3364. [PMID: 37444475 DOI: 10.3390/cancers15133364] [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: 05/26/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The anti-disialoganglioside (GD2) monoclonal antibody dinutuximab beta is approved for the maintenance treatment of high-risk neuroblastoma. Dinutuximab beta combined with different chemotherapy regimens is being investigated in various clinical settings. We conducted a retrospective clinical chart review of 25 patients with relapsed/refractory neuroblastoma who had failed ≥1 second-line therapy and received compassionate use treatment with dinutuximab beta long-term infusion combined with the induction chemotherapy regimens N5 (cisplatin, etoposide, vindesine) and N6 (vincristine, dacarbazine, ifosfamide, doxorubicin) recommended by the German Pediatric Oncology and Hematology Group [GPOH] guidelines. The treatment did not result in any unexpected severe toxicities or in any major treatment delays. Grade 3/4 pain was reported by 4/25 patients in cycle 1, decreasing to 0/9 patients in cycles 3 and 4. The median follow-up was 0.6 years. The best response in this group was 48% (12/25 patients), which included three patients with minor responses. At 1 year, the estimated event-free survival was 27% (95% confidence interval [CI] 8-47) and overall survival was 44% (95% CI 24-65). Combining long-term infusion of dinutuximab beta with N5 and N6 chemotherapy demonstrated an acceptable safety profile and encouraging objective response rates in heavily pretreated patients with high-risk neuroblastoma, warranting further evaluation in clinical trials.
Collapse
Affiliation(s)
- Holger N Lode
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Ruth Ladenstein
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, 1090 Vienna, Austria
- Department for Studies and Statistics and Integrated Research and Project, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung GmbH, 1090 Vienna, Austria
| | - Sascha Troschke-Meurer
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Linda Struppe
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Nikolai Siebert
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Maxi Zumpe
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Karoline Ehlert
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Stefanie Huber
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Evgenia Glogova
- Department of Paediatrics, St. Anna Children's Hospital, Medical University of Vienna, 1090 Vienna, Austria
- Department for Studies and Statistics and Integrated Research and Project, Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung GmbH, 1090 Vienna, Austria
| | | | - Angelika Eggert
- Clinic for Pediatric Hematology and Oncology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Anna Zaniewska-Tekieli
- Department of Pediatric Oncology and Hematology, Jagiellonian University Medical College, 31-008 Krakow, Poland
| | - Walentyna Balwierz
- Department of Pediatric Oncology and Hematology, Jagiellonian University Medical College, 31-008 Krakow, Poland
| | - Aleksandra Wieczorek
- Department of Pediatric Oncology and Hematology, Jagiellonian University Medical College, 31-008 Krakow, Poland
| |
Collapse
|
45
|
Pinto N, Naranjo A, Ding X, Zhang FF, Hibbitts E, Kennedy R, Tibbetts R, Wong-Michalak S, Craig DW, Manojlovic Z, Hogarty MD, Kreissman S, Bagatell R, Irwin MS, Park JR, Asgharzadeh S. Impact of Genomic and Clinical Factors on Outcome of Children ≥18 Months of Age with Stage 3 Neuroblastoma with Unfavorable Histology and without MYCN Amplification: A Children's Oncology Group (COG) Report. Clin Cancer Res 2023; 29:1546-1556. [PMID: 36749880 PMCID: PMC10106446 DOI: 10.1158/1078-0432.ccr-22-3032] [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: 09/30/2022] [Revised: 12/13/2022] [Accepted: 02/06/2023] [Indexed: 02/09/2023]
Abstract
PURPOSE Patients ≥18 months of age with International Neuroblastoma Staging System (INSS) stage 3 unfavorable histology (UH), MYCN-nonamplified (MYCN-NA) tumors have favorable survival rates compared with other high-risk neuroblastoma populations. The impact of select clinical and biological factors on overall survival (OS) and event-free survival (EFS) were evaluated. EXPERIMENTAL DESIGN Patients enrolled on Children's Oncology Group (COG) A3973 (n = 34), ANBL0532 (n = 27), and/or biology protocol ANBL00B1 (n = 72) were analyzed. Tumors with available DNA (n = 65) and RNA (n = 42) were subjected to whole-exome sequencing (WES) and RNA sequencing. WES analyses and gene expression profiling were evaluated for their impact on survival. Multivariate analyses of EFS/OS using significant factors from univariate analyses were performed. RESULTS 5-year EFS/OS for patients treated with high-risk therapy on A3973 and ANBL0532 were 73.0% ± 8.1%/87.9% ± 5.9% and 61.4% ± 10.2%/73.0% ± 9.2%, respectively (P = 0.1286 and P = 0.2180). In the A3973/ANBL0532 cohort, patients with less than partial response (PR; n = 5) at end-induction had poor outcomes (5-year EFS/OS: 0%/20.0% ± 17.9%. Univariate analyses of WES data revealed that subjects whose tumors had chromosome 1p or 11q loss/LOH and chromosome 5 or 9 segmental chromosomal aberrations had inferior EFS compared with those with tumors without these aberrations. Multivariate analysis revealed that 11q loss/LOH was an independent predictor of inferior OS [HR, 3.116 (95% confidence interval, 1.034-9.389), P = 0.0435]. CONCLUSIONS Patients ≥18 months of age at diagnosis who had tumors with UH and MYCN-NA INSS stage 3 neuroblastoma assigned to high-risk therapy had an 81.6% ± 5.3% 5-year OS. Less than PR to induction therapy and chromosome 11q loss/LOH are independent predictors of inferior outcome and identify patients who should be eligible for future high-risk clinical trials.
Collapse
Affiliation(s)
- Navin Pinto
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Arlene Naranjo
- Children’s Oncology Group Data and Statistics Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Xiangming Ding
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Fan F. Zhang
- Children’s Oncology Group Data and Statistics Center, Monrovia, California, USA
| | - Emily Hibbitts
- Children’s Oncology Group Data and Statistics Center, Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Rebekah Kennedy
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Rachelle Tibbetts
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Shannon Wong-Michalak
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - David W Craig
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Zarko Manojlovic
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| | - Michael D. Hogarty
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan Kreissman
- Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Rochelle Bagatell
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meredith S. Irwin
- Department of Pediatrics, University of Toronto, Toronto, Ontario, CANADA
| | - Julie R. Park
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Shahab Asgharzadeh
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
46
|
Desai AV, Elmuti L, Cahaney C, De Guzman RM, Streby KA, Cohn SL. Multimodality treatment for recurrent neuroblastoma in the central nervous system. Pediatr Blood Cancer 2023; 70:e30227. [PMID: 36720647 DOI: 10.1002/pbc.30227] [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: 10/07/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 02/02/2023]
Abstract
Survival for patients with recurrent central nervous system (CNS) neuroblastoma remains poor. A single-institutional study demonstrated the potential of multimodality therapy, including compartmental intrathecal radioimmunotherapy (cRIT) with 131 I-3F8 or 131 I-8H9 to increase the survival of neuroblastoma patients with CNS relapse. However, not all patients are able to receive this therapy. We report three patients with CNS neuroblastoma who remain disease-free 3-9 years after receiving multimodality treatment without cRIT. Additional studies to identify patients most likely to benefit from cRIT are warranted.
Collapse
Affiliation(s)
- Ami V Desai
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Lena Elmuti
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Christine Cahaney
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | | | - Keri A Streby
- Department of Pediatrics, The Ohio State University/Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Susan L Cohn
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
47
|
Vassal G, de Rojas T, Pearson ADJ. Impact of the EU Paediatric Medicine Regulation on new anti-cancer medicines for the treatment of children and adolescents. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:214-222. [PMID: 36682367 DOI: 10.1016/s2352-4642(22)00344-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 01/21/2023]
Abstract
The European Paediatric Medicine Regulation was launched in 2007, aiming to provide better medicines for children. However, its benefit for paediatric patients with cancer has been questioned and the European Paediatric and Orphan Regulations have been under review since November, 2020. To ascertain the effect of the European Paediatric Medicine Regulation, all paediatric anti-cancer medicines assessed by the European Medicines Agency from 1995 to 2022 were identified and reviewed using the agency's public assessment reports, and all Paediatric Investigation Plans granted since 2007 were analysed. 16 new molecular entities (NMEs; ie, a drug that contains an active moiety that had never been approved before) have been approved since the regulation was launched in 2007. The number of paediatric marketing authorisations increased from 2007 but represented the same 17% of all anti-cancer drug marketing authorisations before and after 2007. After 2007, nine (56%) of 16 NMEs were first authorised both in adults and children. For seven NMEs, a first paediatric indication was approved with a median lag time of 6·4 years (range 1·2-21·5 years) after the first authorisation in adults. Half of NMEs were authorised for the treatment of malignancies responsible for only 5·4% of all European childhood cancer deaths, including three medicines for melanoma and thyroid cancer-adult cancers occurring very rarely in children. The increased number of paediatric anti-cancer NMEs after 2007 is a result of the major increase in new medicines authorised for adult cancers since 2005 rather than a direct effect of the Paediatric Regulation. Paediatric development of these NMEs was driven by their adult market and did not address major unmet medical needs of children and adolescents with cancer. An improved, fit-for-purpose regulatory environment that incentivises paediatric drug development based on mechanism of action, better incentives, and a systematic multi-stakeholder engagement, with greater investment from industry, public funding, and non-governmental organisations, will increase the number of new medicines approved in the future to cure more children and adolescents with cancer.
Collapse
Affiliation(s)
- Gilles Vassal
- ACCELERATE, Brussels, Belgium; Department of Paediatric and Adolescent Oncology, Institut Gustave Roussy and Paris-Saclay University, Villejuif, France.
| | | | | |
Collapse
|
48
|
Flaadt T, Ladenstein RL, Ebinger M, Lode HN, Arnardóttir HB, Poetschger U, Schwinger W, Meisel R, Schuster FR, Döring M, Ambros PF, Queudeville M, Fuchs J, Warmann SW, Schäfer J, Seitz C, Schlegel P, Brecht IB, Holzer U, Feuchtinger T, Simon T, Schulte JH, Eggert A, Teltschik HM, Illhardt T, Handgretinger R, Lang P. Anti-GD2 Antibody Dinutuximab Beta and Low-Dose Interleukin 2 After Haploidentical Stem-Cell Transplantation in Patients With Relapsed Neuroblastoma: A Multicenter, Phase I/II Trial. J Clin Oncol 2023:JCO2201630. [PMID: 36854071 DOI: 10.1200/jco.22.01630] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
PURPOSE Patients with relapsed high-risk neuroblastoma (rHR-NB) have a poor prognosis. We hypothesized that graft-versus-neuroblastoma effects could be elicited by transplantation of haploidentical stem cells (haplo-SCT) exploiting cytotoxic functions of natural killer cells and their activation by the anti-GD2 antibody dinutuximab beta (DB). This phase I/II trial assessed safety, feasibility, and outcomes of immunotherapy with DB plus subcutaneous interleukin-2 (scIL2) after haplo-SCT in patients with rHR-NB. METHODS Patients age 1-21 years underwent T-/B-cell-depleted haplo-SCT followed by DB and scIL2. The primary end point 'success of treatment' encompassed patients receiving six cycles, being alive 180 days after end of trial treatment without progressive disease, unacceptable toxicity, acute graft-versus-host-disease (GvHD) ≥grade 3, or extensive chronic GvHD. RESULTS Seventy patients were screened, and 68 were eligible for immunotherapy. Median number of DB cycles was 6 (range, 1-9). Median number of scIL2 cycles was 3 (1-6). The primary end point was met by 37 patients (54.4%). Median observation time was 7.8 years. Five-year event-free survival (EFS) and overall survival from start of trial treatment were 43% (95% CI, 31 to 55) and 53% (95% CI, 41 to 65), respectively. Five-year EFS among patients in complete remission (CR; 52%; 95% CI, 31 to 69) or partial remission (44%; 95% CI, 27 to 60) before immunotherapy were significantly better compared with patients with nonresponse/mixed response/progressive disease (13%; 95% CI, 1 to 42; P = .026). Overall response rate in 43 patients with evidence of disease after haplo-SCT was 51% (22 patients), with 15 achieving CR (35%). Two patients developed GvHD grade 2 and 3 each. No unexpected adverse events occurred. CONCLUSION DB therapy after haplo-SCT in patients with rHR-NB is feasible, with low risk of inducing GvHD, and results in long-term remissions likely attributable to increased antineuroblastoma activity by donor-derived effector cells.
Collapse
Affiliation(s)
- Tim Flaadt
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ruth L Ladenstein
- St Anna Children's Hospital and Children's Cancer Research Institute, Department of Studies and Statistics for Integrated Research and Projects, Medical University of Vienna, Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Martin Ebinger
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Holger N Lode
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Helga Björk Arnardóttir
- Department for Studies and Statistics and Integrated Research, Children's Cancer Research Institute, Vienna, Austria
| | - Ulrike Poetschger
- Department for Studies and Statistics and Integrated Research, Children's Cancer Research Institute, Vienna, Austria
| | - Wolfgang Schwinger
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Friedhelm R Schuster
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Michaela Döring
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Peter F Ambros
- CCRI, Children's Cancer Research Institute, Vienna, Department of Tumor Biology and Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Manon Queudeville
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Steven W Warmann
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Jürgen Schäfer
- Department for Diagnostic and Interventional Radiology, University Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Christian Seitz
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (Exc 2180) "Image-guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Germany
| | - Patrick Schlegel
- Children's Medical Research Institute, The Cancer Centre for Children, The Children's Hospital Westmead, University of Sydney, Sydney, Australia
| | - Ines B Brecht
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ursula Holzer
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Tobias Feuchtinger
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology and Hematology, University Hospital, University of Cologne, Cologne, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology/Hematology, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology/Hematology, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Heiko-Manuel Teltschik
- Department of Hematology and Oncology, Children's Hospital Stuttgart-Olgahospital, Stuttgart, Germany
| | - Toni Illhardt
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (Exc 2180) "Image-guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Germany
| |
Collapse
|
49
|
Wieczorek A, Zaniewska-Tekieli A, Ehlert K, Pawinska-Wasikowska K, Balwierz W, Lode H. Dinutuximab beta combined with chemotherapy in patients with relapsed or refractory neuroblastoma. Front Oncol 2023; 13:1082771. [PMID: 36816982 PMCID: PMC9936065 DOI: 10.3389/fonc.2023.1082771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Prognosis in children with refractory and relapsed high-risk neuroblastoma is poor. Only a minority of patients obtain remission when treated with second-line chemotherapy regimens. Chemotherapy combined with anti-GD2 antibodies has previously been shown to increase response and survival rates. We retrospectively analyzed a cohort of 25 patients with relapsed or refractory high-risk neuroblastoma who were treated with irinotecan/temozolomide chemotherapy in combination with the anti-GD2 antibody dinutuximab beta. The therapy resulted in an objective response rate of 64%, with 32% of patients achieving a complete response. Response to treatment was observed in patients with refractory disease (n=5) and those with first (n=12) or consecutive (n=8) relapses, including patients with progressing disease. In four patients, best response was achieved after more than 5 cycles, suggesting that some patients may benefit from prolonged chemotherapy and dinutuximab beta treatment. Fourteen of our 25 patients had previously received dinutuximab beta, four of whom achieved complete response and six partial response (objective response rate 71%). The therapy was well tolerated, even in heavily pre-treated patients and those who had previously received dinutuximab beta treatment. Toxicities were comparable to those previously reported for the individual therapies, and no discontinuations due to toxicities occurred. Combination of chemotherapy with dinutuximab beta is a promising treatment option for patients with relapsed or refractory high-risk neuroblastoma and should be further explored in clinical studies.
Collapse
Affiliation(s)
- Aleksandra Wieczorek
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Anna Zaniewska-Tekieli
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Karoline Ehlert
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Katarzyna Pawinska-Wasikowska
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Walentyna Balwierz
- Pediatric Oncology and Hematology, Jagiellonian University Medical College, Krakow, Poland
- Pediatric Oncology and Hematology, University Children's Hospital of Krakow, Krakow, Poland
| | - Holger Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
50
|
Erbe AK, Diccianni MB, Mody R, Naranjo A, Zhang FF, Birstler J, Kim K, Feils AS, Hung JT, London WB, Shulkin BL, Mathew V, Parisi MT, Servaes S, Asgharzadeh S, Maris JM, Park J, Yu AL, Sondel PM, Bagatell R. KIR/KIR-ligand genotypes and clinical outcomes following chemoimmunotherapy in patients with relapsed or refractory neuroblastoma: a report from the Children's Oncology Group. J Immunother Cancer 2023; 11:e006530. [PMID: 36822669 PMCID: PMC9950969 DOI: 10.1136/jitc-2022-006530] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND In the Children's Oncology Group ANBL1221 phase 2 trial for patients with first relapse/first declaration of refractory high-risk neuroblastoma, irinotecan and temozolomide (I/T) combined with either temsirolimus (TEMS) or immunotherapy (the anti-GD2 antibody dinutuximab (DIN) and granulocyte macrophage colony stimulating factory (GM-CSF)) was administered. The response rate among patients treated with I/T/DIN/GM-CSF in the initial cohort (n=17) was 53%; additional patients were enrolled to permit further evaluation of this chemoimmunotherapy regimen. Potential associations between immune-related biomarkers and clinical outcomes including response and survival were evaluated. METHODS Patients were evaluated for specific immunogenotypes that influence natural killer (NK) cell activity, including killer immunoglobulin-like receptors (KIRs) and their ligands, Fc gamma receptors, and NCR3. Total white cells and leucocyte subsets were assessed via complete blood counts, and flow cytometry of peripheral blood mononuclear cells was performed to assess the potential association between immune cell subpopulations and surface marker expression and clinical outcomes. Appropriate statistical tests of association were performed. The Bonferroni correction for multiple comparisons was performed where indicated. RESULTS Of the immunogenotypes assessed, the presence or absence of certain KIR and their ligands was associated with clinical outcomes in patients treated with chemoimmunotherapy rather than I/T/TEMS. While median values of CD161, CD56, and KIR differed in responders and non-responders, statistical significance was not maintained in logistic regression models. White cell and neutrophil counts were associated with differences in survival outcomes, however, increases in risk of event in patients assigned to chemoimmunotherapy were not clinically significant. CONCLUSIONS These findings are consistent with those of prior studies showing that KIR/KIR-ligand genotypes are associated with clinical outcomes following anti-GD2 immunotherapy in children with neuroblastoma. The current study confirms the importance of KIR/KIR-ligand genotype in the context of I/T/DIN/GM-CSF chemoimmunotherapy administered to patients with relapsed or refractory disease in a clinical trial. These results are important because this regimen is now widely used for treatment of patients at time of first relapse/first declaration of refractory disease. Efforts to assess the role of NK cells and genes that influence their function in response to immunotherapy are ongoing. TRIAL REGISTRATION NUMBER NCT01767194.
Collapse
Affiliation(s)
- Amy K Erbe
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin, USA
| | - Mitch B Diccianni
- Department of Pediatrics, University of California, San Diego, California, USA
| | - Rajen Mody
- C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Arlene Naranjo
- Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, Florida, USA
| | - Fan F Zhang
- Children's Oncology Group Statistics and Data Center, Monrovia, California, USA
| | - Jen Birstler
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin, USA
| | - KyungMann Kim
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin, USA
| | - Arika S Feils
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin, USA
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wendy B London
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Barry L Shulkin
- Departments of Diagnostic Imaging and Comprehensive Cancer Center, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Varsha Mathew
- Department of Pediatrics, University of California, San Diego, California, USA
| | - Marguerite T Parisi
- Department of Pediatrics, Seattle Children's Hospital and the University, Seattle, Washington, USA
| | - Sabah Servaes
- Department of Pediatrics, The Children's Hospital, Philadelphia, Pennsylvania, USA
| | - Shahab Asgharzadeh
- Department Cancer and Blood Disease Institute, Childrens Hospital of Los Angeles, Los Angeles, California, USA
| | - John M Maris
- Department of Pediatrics, The Children's Hospital, Philadelphia, Pennsylvania, USA
| | - Julie Park
- Department of Pediatrics, Seattle Children's Hospital and the University, Seattle, Washington, USA
| | - Alice L Yu
- Department of Pediatrics, University of California, San Diego, California, USA
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin, USA
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - Rochelle Bagatell
- Department of Pediatrics, The Children's Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|