Skip to main content
Top
Published in: memo - Magazine of European Medical Oncology 3/2021

Open Access 01-09-2021 | Targeted Therapy | short review

Therapy concepts in the context of precision medicine for pediatric malignancies—children are not adults

Authors: Elisabeth Salzer, Caroline Hutter

Published in: memo - Magazine of European Medical Oncology | Issue 3/2021

Login to get access

Summary

Cancer remains the leading cause of death from disease among children beyond the age of one. Survival of pediatric patients with cancer has dramatically improved over the last decades but some tumors remain almost intractable and relapse is still associated with an infaust prognosis. Despite the heterogeneity of pediatric malignancies, most treatments include the same set of generic therapies.  Optimizing delivery of conventional therapeutics has been the driving force behind continuous improvements but further escalation of conventional therapy is unlikely to improve outcomes. The limited success of targeted drugs in pediatric cancer patients, originally developed for cancers in adults, can be connected to the different etiology of tumors in children versus adults. In addition, many pediatric cancers lack reliable biomarkers, cannot be studied in large cohorts and only few available therapies target abberations specific for certain pediatric cancers.
These observations have led to the establishment of pediatric precision-medicine programs. The major goal of these programs is to identify patient-tailored molecular treatment plans that will eventually improve quality of life and survival. Despite the initial euphemism, the impact of actionable matched treatments and the most adequate value-based genomics strategies are not yet well established. A non-competitive collaborative model based on pediatric cancer priorities and strong collaboration between academia, pharmaceutical companies and regulators is needed. In the near future, clinical trials need to focus on biologically defined patient subsets, in an even smaller patient population. A major collaborative effort between all associated groups will be necessary to ensure success of pediatric precision cancer medicine.
Literature
1.
go back to reference Norris RE, Adamson PC. Challenges and opportunities in childhood cancer drug development. Nat Rev Cancer. 2012;12:776–82.CrossRef Norris RE, Adamson PC. Challenges and opportunities in childhood cancer drug development. Nat Rev Cancer. 2012;12:776–82.CrossRef
3.
go back to reference Pritchard-Jones K, Hargrave D. Declining childhood and adolescent cancer mortality: great progress but still much to be done. Cancer. 2014;120:2388–91.CrossRef Pritchard-Jones K, Hargrave D. Declining childhood and adolescent cancer mortality: great progress but still much to be done. Cancer. 2014;120:2388–91.CrossRef
4.
go back to reference Smith MA, et al. Outcomes for children and adolescents with cancer: challenges for the twenty-first century. J Clin Oncol. 2010;28:2625–34.CrossRef Smith MA, et al. Outcomes for children and adolescents with cancer: challenges for the twenty-first century. J Clin Oncol. 2010;28:2625–34.CrossRef
6.
7.
go back to reference Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30:507–19.CrossRef Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30:507–19.CrossRef
8.
9.
go back to reference Rahal Z, Abdulhai F, Kadara H, Saab R. Genomics of adult and pediatric solid tumors. Am J Cancer Res. 2018;8:1356–86.PubMedPubMedCentral Rahal Z, Abdulhai F, Kadara H, Saab R. Genomics of adult and pediatric solid tumors. Am J Cancer Res. 2018;8:1356–86.PubMedPubMedCentral
10.
go back to reference Dharia NV, et al. A first-generation pediatric cancer dependency map. Nat Genet. 2021;53:529–38.CrossRef Dharia NV, et al. A first-generation pediatric cancer dependency map. Nat Genet. 2021;53:529–38.CrossRef
11.
go back to reference Gröbner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555:321–7.CrossRef Gröbner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555:321–7.CrossRef
12.
go back to reference Ma X, et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature. 2018;555:371–6.CrossRef Ma X, et al. Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature. 2018;555:371–6.CrossRef
13.
go back to reference Filbin M, Monje M. Developmental origins and emerging therapeutic opportunities for childhood cancer. Nat Med. 2019;25:367–76.CrossRef Filbin M, Monje M. Developmental origins and emerging therapeutic opportunities for childhood cancer. Nat Med. 2019;25:367–76.CrossRef
14.
go back to reference Li Z, et al. Developmental stage—selective effect of somatically mutated leukemogenic transcription factor GATA1. Nat Genet. 2005;37:613–9.CrossRef Li Z, et al. Developmental stage—selective effect of somatically mutated leukemogenic transcription factor GATA1. Nat Genet. 2005;37:613–9.CrossRef
15.
go back to reference Huntly BJP, et al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cells. 2004;6:587–96.CrossRef Huntly BJP, et al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cells. 2004;6:587–96.CrossRef
16.
go back to reference Funato K, Major T, Lewis PW, Allis CD, Tabar V. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science. 2014;346:1529–33.CrossRef Funato K, Major T, Lewis PW, Allis CD, Tabar V. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science. 2014;346:1529–33.CrossRef
17.
go back to reference Han Z‑Y, et al. The occurrence of intracranial rhabdoid tumours in mice depends on temporal control of Smarcb1 inactivation. Nat Commun. 2016;7:10421.CrossRef Han Z‑Y, et al. The occurrence of intracranial rhabdoid tumours in mice depends on temporal control of Smarcb1 inactivation. Nat Commun. 2016;7:10421.CrossRef
18.
go back to reference Ng JMY, et al. Generation of a mouse model of atypical teratoid/rhabdoid tumor of the central nervous system through combined deletion of Snf5 and p53. Cancer Res. 2015;75:4629–39.CrossRef Ng JMY, et al. Generation of a mouse model of atypical teratoid/rhabdoid tumor of the central nervous system through combined deletion of Snf5 and p53. Cancer Res. 2015;75:4629–39.CrossRef
19.
go back to reference Vitte J, Gao F, Coppola G, Judkins AR, Giovannini M. Timing of Smarcb1 and Nf2 inactivation determines schwannoma versus rhabdoid tumor development. Nat Commun. 2017;8:300.CrossRef Vitte J, Gao F, Coppola G, Judkins AR, Giovannini M. Timing of Smarcb1 and Nf2 inactivation determines schwannoma versus rhabdoid tumor development. Nat Commun. 2017;8:300.CrossRef
20.
go back to reference Vogelstein B, et al. Cancer genome landscapes. Science. 2013;339:1546–58.CrossRef Vogelstein B, et al. Cancer genome landscapes. Science. 2013;339:1546–58.CrossRef
21.
go back to reference Schwartzentruber J, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–31.CrossRef Schwartzentruber J, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–31.CrossRef
22.
go back to reference Burdach SEG, Westhoff M‑A, Steinhauser MF, Debatin K‑M. Precision medicine in pediatric oncology. Mol Cell Pediatr. 2018;5:6.CrossRef Burdach SEG, Westhoff M‑A, Steinhauser MF, Debatin K‑M. Precision medicine in pediatric oncology. Mol Cell Pediatr. 2018;5:6.CrossRef
23.
go back to reference Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372:793–5.CrossRef Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372:793–5.CrossRef
25.
go back to reference van Tilburg CM, et al. The pediatric precision oncology study INFORM: Clinical outcome and benefit for molecular subgroups. J Clin Oncol. 2020;38:LBA10503.CrossRef van Tilburg CM, et al. The pediatric precision oncology study INFORM: Clinical outcome and benefit for molecular subgroups. J Clin Oncol. 2020;38:LBA10503.CrossRef
26.
go back to reference Hadjadj D, Deshmukh S, Jabado N. Entering the era of precision medicine in pediatric oncology. Nat Med. 2020;26:1684–5.CrossRef Hadjadj D, Deshmukh S, Jabado N. Entering the era of precision medicine in pediatric oncology. Nat Med. 2020;26:1684–5.CrossRef
29.
go back to reference Tannock IF, Hickman JA. Limits to personalized cancer medicine. N Engl J Med. 2016;375:1289–94.CrossRef Tannock IF, Hickman JA. Limits to personalized cancer medicine. N Engl J Med. 2016;375:1289–94.CrossRef
30.
go back to reference Vasconcellos VF, Colli LM, Awada A, de Castro G Jr. Precision oncology: as much expectations as limitations. Ecancermedicalscience. 2018;12:ed86.PubMedPubMedCentral Vasconcellos VF, Colli LM, Awada A, de Castro G Jr. Precision oncology: as much expectations as limitations. Ecancermedicalscience. 2018;12:ed86.PubMedPubMedCentral
31.
go back to reference Mody RJ, et al. Integrative clinical sequencing in the management of refractory or relapsed cancer in youth. JAMA. 2015;314:913.CrossRef Mody RJ, et al. Integrative clinical sequencing in the management of refractory or relapsed cancer in youth. JAMA. 2015;314:913.CrossRef
32.
go back to reference Worst BC, et al. Next-generation personalised medicine for high-risk paediatric cancer patients—the INFORM pilot study. Eur J Cancer. 2016;65:91–101.CrossRef Worst BC, et al. Next-generation personalised medicine for high-risk paediatric cancer patients—the INFORM pilot study. Eur J Cancer. 2016;65:91–101.CrossRef
33.
go back to reference Harris MH, et al. Multicenter feasibility study of tumor molecular profiling to inform therapeutic decisions in advanced pediatric solid tumors: the Individualized Cancer Therapy (iCat) study. JAMA Oncol. 2016;2:608.CrossRef Harris MH, et al. Multicenter feasibility study of tumor molecular profiling to inform therapeutic decisions in advanced pediatric solid tumors: the Individualized Cancer Therapy (iCat) study. JAMA Oncol. 2016;2:608.CrossRef
34.
go back to reference Tsoli M, et al. Integration of genomics, high throughput drug screening, and personalized xenograft models as a novel precision medicine paradigm for high risk pediatric cancer. Cancer Biol Ther. 2018;19:1078–87.CrossRef Tsoli M, et al. Integration of genomics, high throughput drug screening, and personalized xenograft models as a novel precision medicine paradigm for high risk pediatric cancer. Cancer Biol Ther. 2018;19:1078–87.CrossRef
35.
go back to reference Li A, Bergan RC. Clinical trial design: past, present, and future in the context of big data and precision medicine. Cancer. 2020;126:4838–46.CrossRef Li A, Bergan RC. Clinical trial design: past, present, and future in the context of big data and precision medicine. Cancer. 2020;126:4838–46.CrossRef
36.
go back to reference Boklan J. Little patients, losing patience: pediatric cancer drug development. Mol Cancer Ther. 2006;5:1905–8.CrossRef Boklan J. Little patients, losing patience: pediatric cancer drug development. Mol Cancer Ther. 2006;5:1905–8.CrossRef
37.
go back to reference Angelini P, Pritchard-Jones K, Hargrave DR. Challenges in incentivizing the pharmaceutical industry to supporting pediatric oncology clinical trials. Clin Investig. 2013;3:101–3.CrossRef Angelini P, Pritchard-Jones K, Hargrave DR. Challenges in incentivizing the pharmaceutical industry to supporting pediatric oncology clinical trials. Clin Investig. 2013;3:101–3.CrossRef
39.
go back to reference Chow EJ, et al. New agents, emerging late effects, and the development of precision survivorship. J Clin Oncol. 2018;36:2231–40.CrossRef Chow EJ, et al. New agents, emerging late effects, and the development of precision survivorship. J Clin Oncol. 2018;36:2231–40.CrossRef
Metadata
Title
Therapy concepts in the context of precision medicine for pediatric malignancies—children are not adults
Authors
Elisabeth Salzer
Caroline Hutter
Publication date
01-09-2021
Publisher
Springer Vienna
Published in
memo - Magazine of European Medical Oncology / Issue 3/2021
Print ISSN: 1865-5041
Electronic ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-021-00743-z

Other articles of this Issue 3/2021

memo - Magazine of European Medical Oncology 3/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine