The European MAPPYACTS Trial: Precision Medicine Program in Pediatric and Adolescent Patients with Recurrent Malignancies

Pablo Berlanga, Gaelle Pierron, Ludovic Lacroix, Mathieu Chicard, Tiphaine Adam de Beaumais, Antonin Marchais, Anne C Harttrampf, Yasmine Iddir, Alicia Larive, Aroa Soriano Fernandez, Imene Hezam, Cecile Chevassus, Virginie Bernard, Sophie Cotteret, Jean-Yves Scoazec, Arnaud Gauthier, Samuel Abbou, Nadege Corradini, Nicolas André, Isabelle Aerts, Estelle Thebaud, Michela Casanova, Cormac Owens, Raquel Hladun-Alvaro, Stefan Michiels, Olivier Delattre, Gilles Vassal, Gudrun Schleiermacher, Birgit Geoerger, Pablo Berlanga, Gaelle Pierron, Ludovic Lacroix, Mathieu Chicard, Tiphaine Adam de Beaumais, Antonin Marchais, Anne C Harttrampf, Yasmine Iddir, Alicia Larive, Aroa Soriano Fernandez, Imene Hezam, Cecile Chevassus, Virginie Bernard, Sophie Cotteret, Jean-Yves Scoazec, Arnaud Gauthier, Samuel Abbou, Nadege Corradini, Nicolas André, Isabelle Aerts, Estelle Thebaud, Michela Casanova, Cormac Owens, Raquel Hladun-Alvaro, Stefan Michiels, Olivier Delattre, Gilles Vassal, Gudrun Schleiermacher, Birgit Geoerger

Abstract

Abstract: MAPPYACTS (NCT02613962) is an international prospective precision medicine trial aiming to define tumor molecular profiles in pediatric patients with recurrent/refractory malignancies in order to suggest the most adapted salvage treatment. From February 2016 to July 2020, 787 patients were included in France, Italy, Ireland, and Spain. At least one genetic alteration leading to a targeted treatment suggestion was identified in 436 patients (69%) with successful sequencing; 10% of these alterations were considered "ready for routine use." Of 356 patients with follow-up beyond 12 months, 107 (30%) received one or more matched targeted therapies-56% of them within early clinical trials-mainly in the AcSé-ESMART platform trial (NCT02813135). Overall, matched treatment resulted in a 17% objective response rate, and of those patients with ready for routine use alterations, it was 38%. In patients with extracerebral tumors, 76% of actionable alterations detected in tumor tissue were also identified in circulating cell-free DNA (cfDNA).

Significance: MAPPYACTS underlines the feasibility of molecular profiling at cancer recurrence in children on a multicenter, international level and demonstrates benefit for patients with selected key drivers. The use of cfDNA deserves validation in prospective studies. Our study highlights the need for innovative therapeutic proof-of-concept trials that address the underlying cancer complexity. This article is highlighted in the In This Issue feature, p. 1171.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Study flow chart. Paired tumor and germline samples were analyzed in all patients with WES. CMTB, clinical molecular tumor board.
Figure 2.
Figure 2.
Oncomap of potentially actionable alterations and canonical fusions in CNS tumors in 118 patients with 119 samples. Tumor types with five or fewer samples are grouped together in “Other.” Only alterations identified five or more times within the whole study are reported here. ATRT, atypical teratoid rhabdoid tumor; EPD, ependymoma; GCT, CNS germ cell tumor; HGG, high-grade glioma; LGG, low-grade glioma; matched, matched therapy received according to recommendations; MB, medulloblastoma; NA, not applicable; NCFUP, follow-up

Figure 3.

Oncomap of potentially actionable alterations…

Figure 3.

Oncomap of potentially actionable alterations and canonical fusions in sarcoma in 213 patients…

Figure 3.
Oncomap of potentially actionable alterations and canonical fusions in sarcoma in 213 patients with 215 samples (A) and other solid tumors in 86 patients with 86 samples (B). Tumor types with five or fewer samples are grouped together in “Other.” Only alterations identified five or more times within the whole study are reported here. aRMS, alveolar rhabdomyosarcoma; CA, carcinoma; DSRCT, desmoplastic small round cell tumor; eRMS, embryonal rhabdomyosarcoma; EWS, Ewing sarcoma; matched, matched therapy received according to recommendations; MPNST, malignant peripheral nerve sheath tumor; NA, not applicable; NBL, neuroblastoma; NCFUP, follow-up < 12 months; NOS, not otherwise specified; NRSTS, non-rhabdomyosarcoma soft-tissue sarcoma; OS, osteosarcoma; Other, other solid tumor; RMS, rhabdomyosarcoma; RT, rhabdoid tumor; SS, synovial sarcoma; subpathology, pathology subtype; US, undifferentiated sarcoma; WT, Wilms tumor.

Figure 4.

Matched targeted treatment recommendations per…

Figure 4.

Matched targeted treatment recommendations per genetic alterations (only one alteration by gene and…

Figure 4.
Matched targeted treatment recommendations per genetic alterations (only one alteration by gene and those occurring in more than five cases are represented). GSi, gamma-secretase inhibitor; PD1/PD1Li, programmed cell death protein 1/ligand immune checkpoint inhibitors; TMB, tumor mutational burden.

Figure 5.

Tumor mutational load in main…

Figure 5.

Tumor mutational load in main cancer types (only disease types with more than…

Figure 5.
Tumor mutational load in main cancer types (only disease types with more than five cases are presented). Median mutational loads and quartiles 25 and 75 are shown as black solid lines for each tumor type. Samples with 2 to 100 ultrahypermutators. ALCL, anaplastic large cell lymphoma; ATRT, atypical teratoid rhabdoid tumor; CA, carcinoma; EPD, ependymoma; EWS, Ewing sarcoma; HB, hepatoblastoma; HGG, high-grade glioma; LGG, low-grade glioma; MB, medulloblastoma; NBL, neuroblastoma; NRSTS, non-RMS soft-tissue sarcoma; OS, osteosarcoma; Other ST, other solid tumors; RMS, rhabdomyosarcoma; WT, Wilms tumor.

Figure 6.

cfDNA and ctDNA ancillary study…

Figure 6.

cfDNA and ctDNA ancillary study in 225 patients with extracranial solid tumors. A,…

Figure 6.
cfDNA and ctDNA ancillary study in 225 patients with extracranial solid tumors. A, cfDNA quantity per type of tumor (ng/mL of plasma, log10 scale). **, indicates statistically significant difference (Wilcoxon test: 3e−04). B, ctDNA fraction calculated by the Facet tool after WES for each type of tumor. ***, indicates statistically significant difference (Wilcoxon test: 1.4e−08). C, Example of a sequencing result for a patient with an alveolar rhabdomyosarcoma. On the top, copy-number profiles generated by the Facet tool, showing the same chromosome alterations identified in the tumor/plasma. Middle, RB1 c.C1333T and TP53 c.A311G mutations were identified in tumor and plasma, whereas TP53 c.A355T was observed only in the plasma. Bottom, Venn diagram of SNVs detected in each type of samples with a good overlap (15 SNVs common between tumor and plasma). D, Oncoprint of SNVs in targeting potentially actionable genes detected in tumor (blue) and plasma (red) for 72 cases with both successful tumor and cfDNA analysis (only cases with at least one SNV in an actionable gene are reported here). Histograms on the top indicate the total number of SNVs in actionable genes in each case (blue, tumor; red, plasma); histograms on the right indicate the percentage of cases with alterations in the indicated actionable gene.
Figure 3.
Figure 3.
Oncomap of potentially actionable alterations and canonical fusions in sarcoma in 213 patients with 215 samples (A) and other solid tumors in 86 patients with 86 samples (B). Tumor types with five or fewer samples are grouped together in “Other.” Only alterations identified five or more times within the whole study are reported here. aRMS, alveolar rhabdomyosarcoma; CA, carcinoma; DSRCT, desmoplastic small round cell tumor; eRMS, embryonal rhabdomyosarcoma; EWS, Ewing sarcoma; matched, matched therapy received according to recommendations; MPNST, malignant peripheral nerve sheath tumor; NA, not applicable; NBL, neuroblastoma; NCFUP, follow-up < 12 months; NOS, not otherwise specified; NRSTS, non-rhabdomyosarcoma soft-tissue sarcoma; OS, osteosarcoma; Other, other solid tumor; RMS, rhabdomyosarcoma; RT, rhabdoid tumor; SS, synovial sarcoma; subpathology, pathology subtype; US, undifferentiated sarcoma; WT, Wilms tumor.
Figure 4.
Figure 4.
Matched targeted treatment recommendations per genetic alterations (only one alteration by gene and those occurring in more than five cases are represented). GSi, gamma-secretase inhibitor; PD1/PD1Li, programmed cell death protein 1/ligand immune checkpoint inhibitors; TMB, tumor mutational burden.
Figure 5.
Figure 5.
Tumor mutational load in main cancer types (only disease types with more than five cases are presented). Median mutational loads and quartiles 25 and 75 are shown as black solid lines for each tumor type. Samples with 2 to 100 ultrahypermutators. ALCL, anaplastic large cell lymphoma; ATRT, atypical teratoid rhabdoid tumor; CA, carcinoma; EPD, ependymoma; EWS, Ewing sarcoma; HB, hepatoblastoma; HGG, high-grade glioma; LGG, low-grade glioma; MB, medulloblastoma; NBL, neuroblastoma; NRSTS, non-RMS soft-tissue sarcoma; OS, osteosarcoma; Other ST, other solid tumors; RMS, rhabdomyosarcoma; WT, Wilms tumor.
Figure 6.
Figure 6.
cfDNA and ctDNA ancillary study in 225 patients with extracranial solid tumors. A, cfDNA quantity per type of tumor (ng/mL of plasma, log10 scale). **, indicates statistically significant difference (Wilcoxon test: 3e−04). B, ctDNA fraction calculated by the Facet tool after WES for each type of tumor. ***, indicates statistically significant difference (Wilcoxon test: 1.4e−08). C, Example of a sequencing result for a patient with an alveolar rhabdomyosarcoma. On the top, copy-number profiles generated by the Facet tool, showing the same chromosome alterations identified in the tumor/plasma. Middle, RB1 c.C1333T and TP53 c.A311G mutations were identified in tumor and plasma, whereas TP53 c.A355T was observed only in the plasma. Bottom, Venn diagram of SNVs detected in each type of samples with a good overlap (15 SNVs common between tumor and plasma). D, Oncoprint of SNVs in targeting potentially actionable genes detected in tumor (blue) and plasma (red) for 72 cases with both successful tumor and cfDNA analysis (only cases with at least one SNV in an actionable gene are reported here). Histograms on the top indicate the total number of SNVs in actionable genes in each case (blue, tumor; red, plasma); histograms on the right indicate the percentage of cases with alterations in the indicated actionable gene.

References

    1. Gatta G, Botta L, Rossi S, Aareleid T, Bielska-Lasota M, Clavel J, et al. . Childhood cancer survival in Europe 1999–2007: results of EUROCARE-5—a population-based study. Lancet Oncol 2014;15:35–47.
    1. Massard C, Michiels S, Ferté C, Le Deley MC, Lacroix L, Hollebecque A, et al. . High-throughput genomics and clinical outcome in hard-to-treat advanced cancers: results of the MOSCATO 01 trial. Cancer Discov 2017;7:586–95.
    1. Le Tourneau C, Delord JP, Gonçalves A, Gavoille C, Dubot C, Isambert N, et al. . Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol 2015;16:1324–34.
    1. Stockley TL, Oza AM, Berman HK, Leighl NB, Knox JJ, Shepherd FA, et al. . Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial. Genome Med 2016;8:109.
    1. Trédan O, Wang Q, Pissaloux D, Cassier P, de la Fouchardière A, Fayette J, et al. . Molecular screening program to select molecular-based recommended therapies for metastatic cancer patients: analysis from the ProfiLER trial. Ann Oncol 2019;30:757–65.
    1. Mossé YP, Voss SD, Lim MS, Rolland D, Minard CG, Fox E, et al. . Targeting ALK with crizotinib in pediatric anaplastic large cell lymphoma and inflammatory myofibroblastic tumor: a Children's Oncology Group Study. J Clin Oncol 2017;35:3215–21.
    1. Geoerger B, Schulte J, Zwaan CM, Casanova M, Fischer M, Moreno L, et al. . Phase I study of ceritinib in pediatric patients (Pts) with malignancies harboring a genetic alteration in ALK (ALK+): safety, pharmacokinetic (PK), and efficacy results. J Clin Oncol 33, 2015(suppl; abstr 10005).
    1. Hargrave DR, Bouffet E, Tabori U, Broniscer A, Cohen KJ, Hansford JR, et al. . Efficacy and safety of dabrafenib in pediatric patients with BRAF V600 mutation–positive relapsed or refractory low-grade glioma: results from a phase I/IIa study. Clin Cancer Res 2019;25:7303–11.
    1. Hargrave DR, Moreno L, Broniscer A, Bouffet E, Aerts I, Andre N, et al. . Dabrafenib in pediatric patients with BRAF V600–positive high-grade glioma (HGG). J Clin Oncol 36, 2018(suppl; abstr 10505).
    1. Fangusaro J, Onar-Thomas A, Young Poussaint T, Wu S, Ligon AH, Lindeman N, et al. . Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol 2019;20:1011–22.
    1. Hong DS, DuBois SG, Kummar S, Farago AF, Albert CM, Rohrberg KS, et al. . Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531–40.
    1. Robinson GW, Gajjar AJ, Gauvain KM, Basu EM, Macy ME, Maese LD, et al. . Phase 1/1B trial to assess the activity of entrectinib in children and adolescents with recurrent or refractory solid tumors including central nervous system (CNS) tumors. J Clin Oncol 37, 2019(suppl; abstr 10009).
    1. Drilon AE, DuBois SG, Farago AF, Geoerger B, Grilley-Olson JE, Hong DS, et al. . Activity of larotrectinib in TRK fusion cancer patients with brain metastases or primary central nervous system tumors. J Clin Oncol 37, 2019(suppl; abstr 2006).
    1. Kieran MW, Chisholm J, Casanova M, Brandes AA, Aerts I, Bouffet E, et al. . Phase I study of oral sonidegib (LDE225) in pediatric brain and solid tumors and a phase II study in children and adults with relapsed medulloblastoma. Neuro Oncol 2017;19:1542–52.
    1. Robinson GW, Orr BA, Wu G, Gururangan S, Lin T, Qaddoumi I, et al. . Vismodegib exerts targeted efficacy against recurrent sonic hedgehog–subgroup medulloblastoma: results from phase II pediatric brain tumor consortium studies PBTC-025B and PBTC-032. J Clin Oncol 2015;33:2646–54.
    1. Vassal G, Faivre L, Geoerger B, Plantaz D, Auvrignon A, Coze C, et al. . Crizotinib in children and adolescents with advanced ROS1, MET, or ALK-rearranged cancer: results of the AcSé phase II trial. J Clin Oncol 34, 2016(suppl; abstr 11509).
    1. Gerdemann U, Lee YA, Henry D, Smith S, Ortiz MV, Rothenberg SM, et al. . First experience of LOXO-292 in the management of pediatric patients with RET-altered cancers. J Clin Oncol 37, 2019(suppl; abstr 10045).
    1. DiNardo CD, Stein EM, de Botton S, Roboz GJ, Altman JK, Mims AS, et al. . Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med 2018;378:2386–98.
    1. Harttrampf AC, Lacroix L, Deloger M, Deschamps F, Puget S, Auger N, et al. . Molecular screening for cancer treatment optimization (MOSCATO-01) in pediatric patients: a single-institutional prospective molecular stratification trial. Clin Cancer Res 2017;23:6101–12.
    1. Pincez T, Clément N, Lapouble E, Pierron G, Kamal M, Bieche I, et al. . Feasibility and clinical integration of molecular profiling for target identification in pediatric solid tumors. Pediatr Blood Cancer 2017;64:e26365.
    1. Mody RJ, Prensner JR, Everett J, Parsons DW, Chinnaiyan AM. Precision medicine in pediatric oncology: lessons learned and next steps. Pediatr Blood Cancer 2017;64:e26288.
    1. Harris MH, DuBois SG, Glade Bender JL, Kim A, Crompton BD, Parker E, 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.
    1. Worst BC, van Tilburg CM, Balasubramanian GP, Fiesel P, Witt R, Freitag A, et al. . Next-generation personalised medicine for high-risk paediatric cancer patients – the INFORM pilot study. Eur J Cancer 2016;65:91–101.
    1. Khater F, Vairy S, Langlois S, Dumoucel S, Sontag T, St-Onge P, et al. . Molecular profiling of hard-to-treat childhood and adolescent cancers. JAMA Netw Open 2019;2:e192906.
    1. Parsons DW, Janeway KA, Patton D, Coffey B, Williams PM, Hamilton SR, et al. . Identification of targetable molecular alterations in the NCI-COG Pediatric MATCH trial. J Clin Oncol 37, 2019(suppl; abstr 10011).
    1. van Tilburg CM, Pfaff E, Pajtler KW, Langenberg KPS, Fiesel P, Jones BC, et al. . The pediatric precision oncology INFORM registry: clinical outcome and benefit for patients with very high-evidence targets. Cancer Discov 2021;11:2764–79.
    1. Kim A, Fox E, Warren K, Blaney SM, Berg SL, Adamson PC, et al. . Characteristics and outcome of pediatric patients enrolled in phase I oncology trials. Oncologist 2008;13:679–89.
    1. Cohen JW, Akshintala S, Kane E, Gnanapragasam H, Widemann BC, Steinberg SM, et al. . A systematic review of pediatric phase I trials in oncology: toxicity and outcomes in the era of targeted therapies. Oncologist 2020;25:532–40.
    1. Wong M, Mayoh C, Lau LMS, Khuong-Quang D-A, Pinese M, Kumar A, et al. . Whole genome, transcriptome and methylome profiling enhances actionable target discovery in high-risk pediatric cancer. Nat Med 2020;26:1742–53.
    1. Pasqualini C, Rubino J, Brard C, Cassard L, André N, Rondof W, et al. . Phase II and biomarker study of programmed cell death protein 1 inhibitor nivolumab and metronomic cyclophosphamide in paediatric relapsed/refractory solid tumours: arm G of AcSé-ESMART, a trial of the European Innovative Therapies for Children with Cancer Consortium. Eur J Cancer 2021;150:53–62.
    1. Bautista F, Paoletti X, Rubino J, Brard C, Rezai K, Nebchi S, et al. . Phase I or II study of ribociclib in combination with topotecan-temozolomide or everolimus in children with advanced malignancies: arms A and B of the AcSé-ESMART Trial. J Clin Oncol 2021; 39:3546–60.
    1. Morscher RJ, Brard C, Berlanga P, Marshall LV, André N, Rubino J, et al. . First in child phase I/II study of the dual mTORC1/2 inhibitor vistusertib (AZD2014) as monotherapy and in combination with topotecan-temozolomide in children with advanced malignancies: Arms E and F of the AcSé-ESMART trial. Eur J Cancer 2021;157:268–77.
    1. Leijen S, van Geel RMJM, Pavlick AC, Tibes R, Rosen L, Razak ARA, et al. . Phase I study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J Clin Oncol 2016;34:4371–80.
    1. Van Tilburg CM, Pfaff E, Pajtler KW, Langenberg K, Fiesel P, Jones BC, et al. . The pediatric precision oncology study INFORM: clinical outcome and benefit for molecular subgroups. J Clin Oncol 38, 2020(suppl; abstr LBA10503).
    1. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov 2022;12:31–46.
    1. Parsons DW, Roy A, Yang Y, Wang T, Scollon S, Bergstrom K, et al. . Diagnostic yield of clinical tumor and germline whole-exome sequencing for children with solid tumors. JAMA Oncol 2016;2:616.
    1. Mody RJ, Wu YM, Lonigro RJ, Cao X, Roychowdhury S, Vats P, et al. . Integrative clinical sequencing in the management of refractory or relapsed cancer in youth. JAMA 2015;314:913.
    1. Zhang J, Walsh MF, Wu G, Edmonson MN, Gruber TA, Easton J, et al. . Germline mutations in predisposition genes in pediatric cancer. N Engl J Med 2015;373:2336–46.
    1. ICGC PedBrain-Seq Project, ICGC MMML-Seq Project, Gröbner SN, Worst BC, Weischenfeldt J, Buchhalter I, et al. . The landscape of geno­mic alterations across childhood cancers. Nature 2018;555:321–7.
    1. Chicard M, Boyault S, Colmet Daage L, Richer W, Gentien D, Pierron G, et al. . Genomic copy number profiling using circulating free tumor DNA highlights heterogeneity in neuroblastoma. Clin Cancer Res 2016;22:5564–73.
    1. Chicard M, Colmet-Daage L, Clement N, Danzon A, Bohec M, Bernard V, et al. . Whole-exome sequencing of cell-free DNA reveals temporo-spatial heterogeneity and identifies treatment-resistant clones in neuroblastoma. Clin Cancer Res 2018;24:939–49.
    1. Escudero L, Llort A, Arias A, Diaz-Navarro A, Martínez-Ricarte F, Rubio-Perez C, et al. . Circulating tumour DNA from the cerebrospinal fluid allows the characterisation and monitoring of medulloblastoma. Nat Commun 2020;11:5376.
    1. Marques Da Costa ME, Daudigeos-Dubus E, Surdez D, Marchais A, Dessen P, Scoazec JY, et al. . Patient-derived xenografts (PDX) development in MAPPYACTS - a pediatric precision cancer medicine trial in relapsed and refractory tumors [abstract]. In: Proceedings of the 51st Congress of the International Society of Paediatric Oncology (SIOP 2019); 2019 Oct 23–26; Lyon, France. Geneva (Switzerland): SIOP; 2019. Abstract nr SIOP19-1241 (oral presentation).
    1. Schulte JH, Moreno L, Ziegler DS, Marshall LV, Zwaan CM, Irwin M, et al. . Final analysis of phase I study of ceritinib in pediatric patients with malignancies harboring activated anaplastic lymphoma kinase (ALK). J Clin Oncol 38, 2020(suppl; abstr 10505).
    1. Ortiz MV, Gerdemann U, Raju SG, Henry D, Smith S, Rothenberg SM, et al. . Activity of the highly specific RET inhibitor selpercatinib (LOXO-292) in pediatric patients with tumors harboring RET gene alterations. JCO Precis Oncol 2020;4:341–7.
    1. Navarrete-Dechent C, Mori S, Barker CA, Dickson MA, Nehal KS. Imatinib treatment for locally advanced or metastatic dermatofibrosarcoma protuberans: a systematic review. JAMA Dermatol 2019;155:361.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47.
    1. Park JR, Bagatell R, Cohn SL, Pearson AD, Villablanca JG, Berthold F, et al. . Revisions to the International Neuroblastoma Response Criteria: a consensus statement from the National Cancer Institute Clinical Trials Planning Meeting. J Clin Oncol 2017;35:2580–7.
    1. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. . Updated response assessment criteria for high-grade gliomas: Response Assessment in Neuro-Oncology Working Group. J Clin Oncol 2010;28:1963–72.

Source: PubMed

3
Suscribir