345 results on '"Zwaan CM"'
Search Results
2. Pharmacokinetics of nilotinib in pediatric patients with Philadelphia chromosome–positive chronic myeloid leukemia or acute lymphoblastic leukemia
- Author
-
Hijiya, N, Michel Zwaan, C, Rizzari, C, Foà, R, Abbink, F, Lancaster, D, Landman-Parker, J, Millot, F, Moppett, J, Nelken, B, Putti, M, Xianbin, T, Sinclair, K, Santanastasio, H, Buchbinder, A, Kearns, P, Michel Zwaan,CM, Putti, MC, Hijiya, N, Michel Zwaan, C, Rizzari, C, Foà, R, Abbink, F, Lancaster, D, Landman-Parker, J, Millot, F, Moppett, J, Nelken, B, Putti, M, Xianbin, T, Sinclair, K, Santanastasio, H, Buchbinder, A, Kearns, P, Michel Zwaan,CM, and Putti, MC
- Abstract
Purpose: We investigated nilotinib exposure in pediatric patients with chronic myeloid leukemia (CML) or Philadelphia chromosome–positive (Phþ) acute lymphoblastic leukemia (ALL) resistant to, relapsed on, refractory to, or intolerant of previous treatment. Patients and Methods: Fifteen patients (aged 1–<18 years) with CML resistant to or intolerant of imatinib and/or dasatinib (n 1⁄4 11) or Phþ ALL relapsed on or refractory to standard therapy (n 1⁄4 4) enrolled in this phase I study. Nilotinib (230 mg/m2 twice daily; equivalent to the adult 400-mg twice-daily dose) was administered orally in 12 or 24 cycles of 28 days. The primary objective was to characterize the pharmacokinetics of nilotinib in pediatric patients. Results: The area under the concentration–time curve at steady state was slightly lower in pediatric patients versus adults (14,751.4 vs. 17,102.9 ng/h/mL); the geometric mean ratio (GMR; pediatric:adult) was 0.86 [90% confidence interval (CI), 0.70–1.06]. Body surface area–adjusted systemic clearance was slightly higher in pediatric versus adult patients (GMR, 1.30; 90% CI, 1.04–1.62). Nilotinib was generally well tolerated. The most common adverse events were headache, vomiting, increased blood bilirubin, and rash. Three patients with CML achieved major molecular response, and three with Phþ ALL achieved complete remission. Conclusions: Nilotinib 230 mg/m2 twice daily in pediatric patients provided a pharmacokinetics and safety profile comparable with the adult reference dose; clinical activity was demonstrated in both CML and Phþ ALL. This dose is recommended for further evaluation in pediatric patients. The safety profile was consistent with that in adults.
- Published
- 2020
3. Low frequency of DNMT3A mutations in pediatric AML, and the identification of the OCI-AML3 cell line as an in vitro model
- Author
-
Hollink, IHIM, Feng, Q, Danen-van Oorschot, AA, Arentsen-Peters, STCJM, Verboon, LJ, Zhang, P, de Haas, V, Reinhardt, D, Creutzig, U, Trka, J, Pieters, R, van den Heuvel-Eibrink, MM, Wang, J, and Zwaan, CM
- Published
- 2012
- Full Text
- View/download PDF
4. The novel calicheamicin-conjugated CD22 antibody inotuzumab ozogamicin (CMC-544) effectively kills primary pediatric acute lymphoblastic leukemia cells
- Author
-
de Vries, JF, Zwaan, CM, De Bie, M, Voerman, JSA, den Boer, ML, van Dongen, JJM, and van der Velden, VHJ
- Published
- 2012
- Full Text
- View/download PDF
5. Favorable prognostic impact of NPM1 gene mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically normal AML
- Author
-
Hollink, IHIM, Zwaan, CM, Zimmermann, M, Arentsen-Peters, TCJM, Pieters, R, Cloos, J, Kaspers, GJL, de Graaf, SSN, Harbott, J, Creutzig, U, Reinhardt, D, van den Heuvel-Eibrink, MM, and Thiede, C
- Published
- 2009
6. Optimal immunocytochemical and flow cytometric detection of P-gp, MRP and LRP in childhood acute lymphoblastic leukemia
- Author
-
Den Boer, ML, Zwaan, CM, Pieters, R, Kazemier, KM, Rottier, MMA, Flens, MJ, Scheper, RJ, and Veerman, AJP
- Published
- 1997
- Full Text
- View/download PDF
7. A phase 1/2, open-label, dose-escalation study of midostaurin in children with relapsed or refractory acute leukaemia
- Author
-
Zwaan, C, Söderhäll, S, Brethon, B, Luciani, M, Rizzari, C, Stam, R, Besse, E, Dutreix, C, Fagioli, F, Ho, P, Dufour, C, Pieters, R, Zwaan, CM, Stam, RW, Ho, PA, Zwaan, C, Söderhäll, S, Brethon, B, Luciani, M, Rizzari, C, Stam, R, Besse, E, Dutreix, C, Fagioli, F, Ho, P, Dufour, C, Pieters, R, Zwaan, CM, Stam, RW, and Ho, PA
- Published
- 2019
8. The genetic basis and cell of origin of mixed phenotype acute leukaemia.
- Author
-
Alexander, TB, Gu, Z, Iacobucci, I, Dickerson, K, Choi, JK, Xu, B, Payne-Turner, D, Yoshihara, H, Loh, ML, Horan, J, Buldini, B, Basso, G, Elitzur, S, de Haas, V, Zwaan, CM, Yeoh, A, Reinhardt, D, Tomizawa, D, Kiyokawa, N, Lammens, T, De Moerloose, B, Catchpoole, D, Hori, H, Moorman, A, Moore, AS, Hrusak, O, Meshinchi, S, Orgel, E, Devidas, M, Borowitz, M, Wood, B, Heerema, NA, Carrol, A, Yang, Y-L, Smith, MA, Davidsen, TM, Hermida, LC, Gesuwan, P, Marra, MA, Ma, Y, Mungall, AJ, Moore, RA, Jones, SJM, Valentine, M, Janke, LJ, Rubnitz, JE, Pui, C-H, Ding, L, Liu, Y, Zhang, J, Nichols, KE, Downing, JR, Cao, X, Shi, L, Pounds, S, Newman, S, Pei, D, Guidry Auvil, JM, Gerhard, DS, Hunger, SP, Inaba, H, Mullighan, CG, Alexander, TB, Gu, Z, Iacobucci, I, Dickerson, K, Choi, JK, Xu, B, Payne-Turner, D, Yoshihara, H, Loh, ML, Horan, J, Buldini, B, Basso, G, Elitzur, S, de Haas, V, Zwaan, CM, Yeoh, A, Reinhardt, D, Tomizawa, D, Kiyokawa, N, Lammens, T, De Moerloose, B, Catchpoole, D, Hori, H, Moorman, A, Moore, AS, Hrusak, O, Meshinchi, S, Orgel, E, Devidas, M, Borowitz, M, Wood, B, Heerema, NA, Carrol, A, Yang, Y-L, Smith, MA, Davidsen, TM, Hermida, LC, Gesuwan, P, Marra, MA, Ma, Y, Mungall, AJ, Moore, RA, Jones, SJM, Valentine, M, Janke, LJ, Rubnitz, JE, Pui, C-H, Ding, L, Liu, Y, Zhang, J, Nichols, KE, Downing, JR, Cao, X, Shi, L, Pounds, S, Newman, S, Pei, D, Guidry Auvil, JM, Gerhard, DS, Hunger, SP, Inaba, H, and Mullighan, CG
- Abstract
Mixed phenotype acute leukaemia (MPAL) is a high-risk subtype of leukaemia with myeloid and lymphoid features, limited genetic characterization, and a lack of consensus regarding appropriate therapy. Here we show that the two principal subtypes of MPAL, T/myeloid (T/M) and B/myeloid (B/M), are genetically distinct. Rearrangement of ZNF384 is common in B/M MPAL, and biallelic WT1 alterations are common in T/M MPAL, which shares genomic features with early T-cell precursor acute lymphoblastic leukaemia. We show that the intratumoral immunophenotypic heterogeneity characteristic of MPAL is independent of somatic genetic variation, that founding lesions arise in primitive haematopoietic progenitors, and that individual phenotypic subpopulations can reconstitute the immunophenotypic diversity in vivo. These findings indicate that the cell of origin and founding lesions, rather than an accumulation of distinct genomic alterations, prime tumour cells for lineage promiscuity. Moreover, these findings position MPAL in the spectrum of immature leukaemias and provide a genetically informed framework for future clinical trials of potential treatments for MPAL.
- Published
- 2018
9. Normal karyotype is a poor prognostic factor in myeloid leukemia of Down syndrome: a retrospective, international study
- Author
-
Blink, Marjolein, Zimmermann, M, von Neuhoff, C, Reinhardt, D, de Haas, V, Hasle, H, O'Brien, MM, Stark, B, Tandonnet, J, Pession, A, Tousovska, K, Cheuk, DKL, Kudo, K, Taga, T, Rubnitz, JE, Haltrich, I, Balwierz, W, Pieters, Rob, Forestier, E, Johansson, B, Eibrink, Marry, Zwaan, CM, Blink, Marjolein, Zimmermann, M, von Neuhoff, C, Reinhardt, D, de Haas, V, Hasle, H, O'Brien, MM, Stark, B, Tandonnet, J, Pession, A, Tousovska, K, Cheuk, DKL, Kudo, K, Taga, T, Rubnitz, JE, Haltrich, I, Balwierz, W, Pieters, Rob, Forestier, E, Johansson, B, Eibrink, Marry, and Zwaan, CM
- Published
- 2014
10. t(6;9)(p22; q34)/DEK-NUP214-rearranged pediatric myeloid leukemia: an international study of 62 patients
- Author
-
Sandahl, JD, Coenen, Eva, Forestier, E, Harbott, J, Johansson, B, Kerndrup, G, Adachi, S, Auvrignon, A, Beverloo, Berna, Cayuela, JM, Chilton, L, Fornerod, M, de Haas, V, Harrison, CJ, Inaba, H, Kaspers, GJL, Liang, DC, Locatelli, F (Franco), Masetti, R, Perot, C, Raimondi, SC, Reinhardt, K, Tomizawa, D, von Neuhoff, N, Zecca, M, Zwaan, CM, Eibrink, Marry, Hasle, H, Sandahl, JD, Coenen, Eva, Forestier, E, Harbott, J, Johansson, B, Kerndrup, G, Adachi, S, Auvrignon, A, Beverloo, Berna, Cayuela, JM, Chilton, L, Fornerod, M, de Haas, V, Harrison, CJ, Inaba, H, Kaspers, GJL, Liang, DC, Locatelli, F (Franco), Masetti, R, Perot, C, Raimondi, SC, Reinhardt, K, Tomizawa, D, von Neuhoff, N, Zecca, M, Zwaan, CM, Eibrink, Marry, and Hasle, H
- Published
- 2014
11. Acute lymphoblastic leukemia in children with Down syndrome: A retrospective analysis from the Ponte di Legno study group
- Author
-
Buitenkamp, T, Izraeli, S, Zimmermann, M, Forestier, E, Heerema, N, van den Heuvel Eibrink, M, Pieters, R, Korbijn, C, Silverman, L, Schmiegelow, K, Liang, D, Horibe, K, Arico, M, Biondi, A, Basso, G, Rabin, K, Schrappe, M, Cario, G, Mann, G, Morak, M, Panzer Grümayer, R, Mondelaers, V, Lammens, T, Cavé, H, Stark, B, Ganmore, I, Moorman, A, Vora, A, Hunger, S, Pui, C, Mullighan, C, Manabe, A, Escherich, G, Kowalczyk, J, Whitlock, J, Zwaan, C, Buitenkamp, TD, Heerema, NA, van den Heuvel Eibrink, MM, Korbijn, CM, Silverman, LB, Liang, DC, Rabin, KR, Moorman, AV, Hunger, SP, Pui, CH, Mullighan, CG, Kowalczyk, JR, Whitlock, JA, Zwaan, CM, BIONDI, ANDREA, Buitenkamp, T, Izraeli, S, Zimmermann, M, Forestier, E, Heerema, N, van den Heuvel Eibrink, M, Pieters, R, Korbijn, C, Silverman, L, Schmiegelow, K, Liang, D, Horibe, K, Arico, M, Biondi, A, Basso, G, Rabin, K, Schrappe, M, Cario, G, Mann, G, Morak, M, Panzer Grümayer, R, Mondelaers, V, Lammens, T, Cavé, H, Stark, B, Ganmore, I, Moorman, A, Vora, A, Hunger, S, Pui, C, Mullighan, C, Manabe, A, Escherich, G, Kowalczyk, J, Whitlock, J, Zwaan, C, Buitenkamp, TD, Heerema, NA, van den Heuvel Eibrink, MM, Korbijn, CM, Silverman, LB, Liang, DC, Rabin, KR, Moorman, AV, Hunger, SP, Pui, CH, Mullighan, CG, Kowalczyk, JR, Whitlock, JA, Zwaan, CM, and BIONDI, ANDREA
- Abstract
Children with Down syndrome (DS) have an increased risk of B-cell precursor (BCP) acute lymphoblastic leukemia (ALL). The prognostic factors and outcome of DS-ALL patients treated in contemporary protocols are uncertain. We studied 653 DS-ALL patients enrolled in 16 international trials from 1995 to 2004. Non-DS BCP-ALL patients from the Dutch Child Oncology Group and Berlin-Frankfurt- Münster were reference cohorts. DS-ALL patients had a higher 8-year cumulative incidence of relapse (26% ± 2% vs 15% ± 1%, P < .001) and 2-year treatment-related mortality (TRM) (7% ± 1% vs 2.0% ± <1%, P < .0001) than non-DS patients, resulting in lower 8-year event-free survival (EFS) (64% ± 2% vs 81% ± 2%, P < .0001) and overall survival (74% ± 2% vs 89% ± 1%, P < .0001). Independent favorable prognostic factors include age <6 years (hazard ratio [HR] = 0.58, P = .002), white blood cell (WBC) count <10 3 109/L (HR = 0.60, P = .005), and ETV6-RUNX1 (HR = 0.14, P = .006) for EFS and age (HR = 0.48, P < .001), ETV6-RUNX1 (HR = 0.1, P = .016) and high hyperdiploidy (HeH) (HR = 0.29, P = .04) for relapse-free survival. TRM was the major cause of death in ETV6-RUNX1 and HeH DSALLs. Thus, while relapse is the main contributor to poorer survival in DS-ALL, infection-associated TRM was increased in all protocol elements, unrelated to treatment phase or regimen. Future strategies to improve outcome in DS-ALL should include improved supportive care throughout therapy and reduction of therapy in newly identified good-prognosis subgroups. (Blood. 2014; 123(1):70-77). © 2014 by The American Society of Hematology.
- Published
- 2014
12. High frequency of copy number variations in Myeloid Leukemia og Down syndrome
- Author
-
Blink, M, van den Heuvel-Eibrink, MM, Balgobind, B, Hollink, IHIM, de Haas, V, Kaspers, GJ, Reinhardt, D, Hasle, Henrik, and Zwaan, CM
- Published
- 2009
13. Identification of novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia
- Author
-
Balgobind, BV, Raimondi, SC, Harbott, J, Zimmermann, M, Alonzo, TA, Auvrignon, A, Beverloo, HB, Chang, M, Creutzig, U, Dworzak, MN, Forestier, E, Gibson, B, Hasle, Henrik, Harrison, CJ, Heerema, NA, Kaspers, GJ, Leszl, A, Litvinko, N, Nigro, LL, Morimoto, A, Perot, C, Pieters, R, Reinhardt, D, Rubnitz, JE, Smith, FO, Stary, J, Stasevich, I, Strehl, S, Taga, T, Tomizawa, D, Webb, D, Zemanova, Z, Zwaan, CM, and van den Heuvel-Eibrink, MM
- Published
- 2009
14. The MLL recombinome of acute leukemias in 2013
- Author
-
Meyer, Claus, Hofmann, J, Burmeister, Thomas, Gröger, D, Park, TS, Emerenciano, M, Pombo de Oliveira, M, Renneville, A, Macintyre, E, Cavé, H, Clappier, E, Mass-Malo, K, Zuna, J, Trka, J, De Braekeleer, E, De Braekeleer, M, Oh, SH, Tsaur, G, Fechina, L, van der Velden, VH, van Dongen, JJ, Delabesse, E, Binato, R, Silva, ML, Kustanovich, A, Aleinikova, O, Harris, MH, Lund-Aho, T, Juvonen, V, Heidenreich, Olaf Torben, Vormoor, J, Choi, WW, Jarosova, M, Kolenova, A, Bueno, C, Menendez, P, Wehner, S, Eckert, C, Talmant, P, Tondeur, S, Lippert, E, Launay, E, Henry, C, Ballerini, P, Lapillone, H, Callanan, MB, Cayuela, JM, Herbaux, C, Cazzaniga, G, Kakadiya, PM, Bohlander, Stefan Klaus, Ahlmann, M, Choi, JR, Gameiro, P, Lee, DS, Krauter, J, Cornillet-Lefebvre, P, Te Kronnie, G, Schäfer, BW, Kubetzko, S, Alonso, CN, zur Stadt, U, Sutton, R, Venn, NC, Izraeli, S, Trakhtenbrot, L, Madsen, HO, Archer, P, Hancock, J, Cerveira, N, Teixeira, MR, Lo Nigro, L, Möricke, A, Stanulla, M, Schrappe, M, Sedék, L, Szczepański, T, Zwaan, CM, Coenen, EA, van den Heuvel-Eibrink, MM, Strehl, S, Dworzak, Michael N., Panzer-Grümayer, Renate, Dingermann, Theodor, Klingebiel, Thomas, Marschalek, Rolf, Meyer, Claus, Hofmann, J, Burmeister, Thomas, Gröger, D, Park, TS, Emerenciano, M, Pombo de Oliveira, M, Renneville, A, Macintyre, E, Cavé, H, Clappier, E, Mass-Malo, K, Zuna, J, Trka, J, De Braekeleer, E, De Braekeleer, M, Oh, SH, Tsaur, G, Fechina, L, van der Velden, VH, van Dongen, JJ, Delabesse, E, Binato, R, Silva, ML, Kustanovich, A, Aleinikova, O, Harris, MH, Lund-Aho, T, Juvonen, V, Heidenreich, Olaf Torben, Vormoor, J, Choi, WW, Jarosova, M, Kolenova, A, Bueno, C, Menendez, P, Wehner, S, Eckert, C, Talmant, P, Tondeur, S, Lippert, E, Launay, E, Henry, C, Ballerini, P, Lapillone, H, Callanan, MB, Cayuela, JM, Herbaux, C, Cazzaniga, G, Kakadiya, PM, Bohlander, Stefan Klaus, Ahlmann, M, Choi, JR, Gameiro, P, Lee, DS, Krauter, J, Cornillet-Lefebvre, P, Te Kronnie, G, Schäfer, BW, Kubetzko, S, Alonso, CN, zur Stadt, U, Sutton, R, Venn, NC, Izraeli, S, Trakhtenbrot, L, Madsen, HO, Archer, P, Hancock, J, Cerveira, N, Teixeira, MR, Lo Nigro, L, Möricke, A, Stanulla, M, Schrappe, M, Sedék, L, Szczepański, T, Zwaan, CM, Coenen, EA, van den Heuvel-Eibrink, MM, Strehl, S, Dworzak, Michael N., Panzer-Grümayer, Renate, Dingermann, Theodor, Klingebiel, Thomas, and Marschalek, Rolf
- Abstract
Chromosomal rearrangements of the human MLL (mixed lineage leukemia) gene are associated with high-risk infant, pediatric, adult and therapy-induced acute leukemias. We used long-distance inverse-polymerase chain reaction to characterize the chromosomal rearrangement of individual acute leukemia patients. We present data of the molecular characterization of 1590 MLL-rearranged biopsy samples obtained from acute leukemia patients. The precise localization of genomic breakpoints within the MLL gene and the involved translocation partner genes (TPGs) were determined and novel TPGs identified. All patients were classified according to their gender (852 females and 745 males), age at diagnosis (558 infant, 416 pediatric and 616 adult leukemia patients) and other clinical criteria. Combined data of our study and recently published data revealed a total of 121 different MLL rearrangements, of which 79 TPGs are now characterized at the molecular level. However, only seven rearrangements seem to be predominantly associated with illegitimate recombinations of the MLL gene (~ 90%): AFF1/AF4, MLLT3/AF9, MLLT1/ENL, MLLT10/AF10, ELL, partial tandem duplications (MLL PTDs) and MLLT4/AF6, respectively. The MLL breakpoint distributions for all clinical relevant subtypes (gender, disease type, age at diagnosis, reciprocal, complex and therapy-induced translocations) are presented. Finally, we present the extending network of reciprocal MLL fusions deriving from complex rearrangements.
- Published
- 2013
15. Different drug sensisivity profiles of acute myeloid and lymphoblastic leukemia and normal peripheral blood mononuclear cells in children with and without Down
- Author
-
Zwaan, CM, Kaspers, GJL, Pieters, Rob, Hählen, K, Janka-Schaub, GE, van Zantwijk, CH, Huismans, DR, Vries, Esther, Rots, MG, Peters, GJ, Jansen, G, Creutzig, U, Veerman, AJP, Pediatrics, and Public Health
- Published
- 2002
16. Wanneer welke diagnostiek of therapie bij anemie en trombopenie?
- Author
-
Appel, IM, Zwaan, CM, Pieters, R., Caron, H.N., and Pediatrics
- Published
- 2000
17. Phase II study of gemcitabine combined with oxaliplatin in relapsed or refractory paediatric solid malignancies: An innovative therapy for children with Cancer European Consortium Study.
- Author
-
Geoerger, B, Chisholm, J, Le Deley, Mc, Gentet, Jc, Zwaan, Cm, Dias, N, Jaspan, T, Hugh, K, Couanet, D, Hain, S, Devos, A, Riccardi, Riccardo, Cesare, C, Boos, J, Frappaz, D, Leblond, P, Aerts, I, Vassal, G., Riccardi, Riccardo (ORCID:0000-0001-7515-6622), Geoerger, B, Chisholm, J, Le Deley, Mc, Gentet, Jc, Zwaan, Cm, Dias, N, Jaspan, T, Hugh, K, Couanet, D, Hain, S, Devos, A, Riccardi, Riccardo, Cesare, C, Boos, J, Frappaz, D, Leblond, P, Aerts, I, Vassal, G., and Riccardi, Riccardo (ORCID:0000-0001-7515-6622)
- Abstract
AIM: To assess objective response rates after 4 cycles of gemcitabine in combination with oxaliplatin in children and adolescents with relapsed or refractory solid tumours. METHODS: This multicentre, non-randomised Phase II study included five strata: neuroblastoma, osteosarcoma, medulloblastoma and other CNS tumours strata with two-stage Simon designs and a miscellaneous, extra-cranial solid tumour stratum with descriptive design. Eligibility criteria included: age 6 months to 21 years; measurable, relapsed or refractory solid malignancy; no more than one previous salvage therapy. Gemcitabine was administered intravenously at 1000 mg/m(2) over 100 min followed by oxaliplatin at 100mg/m(2) over 120 min on Day 1 of a 14-d cycle. Tumour response was assessed every 4 cycles according to WHO criteria. RESULTS: Ninety-three out of 95 patients enrolled in 25 centres received treatment: 12 neuroblastoma; 12 osteosarcoma; 14 medulloblastoma; 13 other CNS tumours and 42 miscellaneous non-CNS solid tumours. Median age was 11.7 years (range, 1.3-20.8 years). Tumour control (CR+PR+SD) at 4 cycles was obtained in 30/93 evaluable patients (32.3%; 95% confidence interval (CI), 22.9-42.7%), including four PR: 1/12 patients with osteosarcoma, 1/12 with medulloblastoma, 1/12 with rhabdomyosarcoma and 1/4 with other sarcoma. Five out of 12 eligible patients with neuroblastoma experienced stable disease. During a total of 481 treatment cycles (median 4, range 1-24 per patient), the most common treatment-related toxicities were haematologic (leukopenia, neutropenia, thrombocytopenia) and neurological (dysesthesia, paresthesia). CONCLUDING STATEMENT: The gemcitabine-oxaliplatin combination administered in a bi-weekly schedule has acceptable safety profile with limited activity in children with relapsed or refractory solid tumours.
- Published
- 2011
18. Toxicity assessment of molecularly targeted drugs incorporated into multiagent chemotherapy regimens for pediatric acute lymphocytic leukemia (ALL): review from an international consensus conference
- Author
-
Horton, T, Sposto, R, Brown, P, Reynolds, C, Hunger, S, Winick, N, Raetz, E, Carroll, W, Arceci, R, Borowitz, M, Gaynon, P, Gore, L, Jeha, S, Maurer, B, Siegel, S, Biondi, A, Kearns, P, Narendran, A, Silverman, L, Smith, M, Zwaan, C, Whitlock, J, Horton, TM, Reynolds, CP, Hunger, SP, Winick, NJ, Raetz, EA, Carroll, WL, Arceci, RJ, Borowitz, MJ, Gaynon, PS, Maurer, BJ, Siegel, SE, BIONDI, ANDREA, Kearns, PR, Silverman, LB, Smith, MA, Zwaan, CM, Whitlock, JA, Horton, T, Sposto, R, Brown, P, Reynolds, C, Hunger, S, Winick, N, Raetz, E, Carroll, W, Arceci, R, Borowitz, M, Gaynon, P, Gore, L, Jeha, S, Maurer, B, Siegel, S, Biondi, A, Kearns, P, Narendran, A, Silverman, L, Smith, M, Zwaan, C, Whitlock, J, Horton, TM, Reynolds, CP, Hunger, SP, Winick, NJ, Raetz, EA, Carroll, WL, Arceci, RJ, Borowitz, MJ, Gaynon, PS, Maurer, BJ, Siegel, SE, BIONDI, ANDREA, Kearns, PR, Silverman, LB, Smith, MA, Zwaan, CM, and Whitlock, JA
- Abstract
One of the challenges of incorporating molecularly targeted drugs into multi-agent chemotherapy (backbone) regimens is defining dose-limiting toxicities (DLTs) of the targeted agent against the background of toxicities of the backbone regimen. An international panel of 22 pediatric acute lymphocytic leukemia (ALL) experts addressed this issue (www.ALLNA.org). Two major questions surrounding DLT assessment were explored: (1) how toxicities can be best defined, assessed, and attributed; and (2) how effective dosing of new agents incorporated into multi-agent ALL clinical trials can be safely established in the face of disease- and therapy-related systemic toxicities. The consensus DLT definition incorporates tolerance of resolving Grade 3 and some resolving Grade 4 toxicities with stringent safety monitoring. This functional DLT definition is being tested in two Children's Oncology Group (COG) ALL clinical trials. © 2010 Wiley-Liss, Inc.
- Published
- 2010
19. The role of the Innovative Therapies for Children with Cancer' (ITCC) European consortium
- Author
-
Zwaan, Cm, Kearns, P, Caron, H, Verschuur, A, Riccardi, Riccardo, Boos, J, Doz, F, Geoerger, B, Morland, B, Vassal, G., Riccardi, Riccardo (ORCID:0000-0001-7515-6622), Zwaan, Cm, Kearns, P, Caron, H, Verschuur, A, Riccardi, Riccardo, Boos, J, Doz, F, Geoerger, B, Morland, B, Vassal, G., and Riccardi, Riccardo (ORCID:0000-0001-7515-6622)
- Abstract
Overall survival from childhood malignancies has dramatically improved, with survival rates now reaching over 70%. Nevertheless, some types of childhood cancer remain a difficult challenge, and for those who survive the burden of treatment can be considerable. The current paradigm for new cancer therapies is to increase our knowledge of the molecular basis of carcinogenesis, followed by the development of cancer-cell specific therapies. Historically, drug development was focused on adult cancers, and the potential efficacy in childhood malignancies was not considered. Recently, a European academic consortium was established, namely 'innovative therapies for children with cancer' (ITCC), to address this unmet need. This initiative is focused on the evaluation of novel agents in pediatric cancer pre-clinical models, and early clinical development of promising new drugs. The number of pediatric patients eligible to participate in such trials is limited, and accurate pre-clinical evaluation may provide evidence-based prioritization for clinical development. Until recently, clinical development of new drugs in childhood cancer was restricted by the limited accessibility of such agents. Recent changes in EU legislation oblige pharmaceutical companies to provide pediatric clinical data for all new drugs relevant to children, including anti-cancer drugs. Pediatric consortiums like ITCC have established networks of expertise with the specific aim of evaluating new drugs for the treatment of childhood cancers. Through proper evaluation in collaborative clinical trials we will learn how best to use these new therapeutic approaches and improve the survival rates and reduce toxicity for children with cancer.
- Published
- 2010
20. Standardization of WT1 mRNA quantitation for minimal residual disease monitoring in childhood AML and implications of WT1 gene mutations: a European multicenter study
- Author
-
Willasch, A, Gruhn, B, Coliva, T, Kalinova, M, Schneider, G, Kreyenberg, H, Steinbach, D, Weber, G, Hollink, I, Zwaan, C, Biondi, A, van der Velden, V, Reinhardt, D, Cazzaniga, G, Bader, P, Trka, J, Willasch, AM, COLIVA, TIZIANA ANGELA, Hollink, IHIM, Zwaan, CM, van der Velden, VHJ, Trka, J., BIONDI, ANDREA, Willasch, A, Gruhn, B, Coliva, T, Kalinova, M, Schneider, G, Kreyenberg, H, Steinbach, D, Weber, G, Hollink, I, Zwaan, C, Biondi, A, van der Velden, V, Reinhardt, D, Cazzaniga, G, Bader, P, Trka, J, Willasch, AM, COLIVA, TIZIANA ANGELA, Hollink, IHIM, Zwaan, CM, van der Velden, VHJ, Trka, J., and BIONDI, ANDREA
- Abstract
A standardized, sensitive and universal method for minimal residual disease (MRD) detection in acute myeloid leukemia (AML) is still pending. Although hyperexpression of Wilms' tumor (WT1) gene transcript has been frequently proposed as an MRD marker in AML, wide comparability of the various methods used for evaluating WT1 expression has not been given. We established and standardized a multicenter approach for quantifying WT1 expression by quantitative reverse transcriptase PCR (qRT-PCR), on the basis of a primer/probe set combination at exons 6 and 7. In a series of quality-control rounds, we analyzed 69 childhood AML samples and 47 normal bone marrow (BM) samples from 4 participating centers. Differences in the individual WT1 expressions levels ranged within <0.5 log of the mean in 82% of the cases. In AML samples, the median WT1/1E+04 Abelson (ABL) expression was 3.5E+03 compared with that of 2.3E+01 in healthy BM samples. As 11.5% of childhood AML samples in this cohort harbored WT1 mutations in exon 7, the effect of mutations on WT1 expression has been investigated, showing that mutated cases expressed significantly higher WT1 levels than wild-type cases. Hence, our approach showed high reproducibility and applicability, even in patients with WT1 mutations; therefore, it can be widely used for the quantitation of WT1 expression in future clinical trials.
- Published
- 2009
21. t(3;11)(q13.13;q23) MLL/KIAA1524
- Author
-
Coenen, EA, primary, Zwaan, CM, additional, van, den Heuvel-Eibrink MM, additional, Beverloo, HB, additional, Marschalek, R, additional, and Meyer, C, additional
- Published
- 2012
- Full Text
- View/download PDF
22. 11q23 rearrangements in de novo childhood acute myeloid leukemia
- Author
-
Coenen, EA, primary, Harbott, J, additional, Zwaan, CM, additional, Raimondi, SC, additional, and van, den Heuvel-Eibrink MM, additional
- Published
- 2012
- Full Text
- View/download PDF
23. First clinical experiences with Gemtuzumab ozogamicin (GO, Mylotarg (c)) in CD33 positive relapsed/refractory leukemia in children
- Author
-
Zwaan, CM, Reinhardt, D, Corbacioglu, S, Jurgens, H, Samuelsson, Ulf, Biondi, A, Smith, OP, Bokkerink, JPM, Tissing, WJE, Creutzig, U, Kaspers, GJL, Zwaan, CM, Reinhardt, D, Corbacioglu, S, Jurgens, H, Samuelsson, Ulf, Biondi, A, Smith, OP, Bokkerink, JPM, Tissing, WJE, Creutzig, U, and Kaspers, GJL
- Published
- 2003
24. Multimodal treatment, including interferon beta, of nasopharyngeal carcinoma in children and young adults: preliminary results from the prospective, multicenter study NPC-2003-GPOH/DCOG.
- Author
-
Buehrlen M, Zwaan CM, Granzen B, Lassay L, Deutz P, Vorwerk P, Staatz G, Gademann G, Christiansen H, Oldenburger F, Tamm M, Mertens R, Buehrlen, Martina, Zwaan, Christian Michel, Granzen, Bernd, Lassay, Lisa, Deutz, Peter, Vorwerk, Peter, Staatz, Gundula, and Gademann, Günther
- Abstract
Background: The authors report preliminary results from a prospective multicenter study (Nasopharyngeal Carcinoma [NPC] 2003 German Society of Pediatric Oncology and Hematology/German Children's Oncology Group [NPC-2003-GPOH/DCOG]).Methods: From 2003 to 2010, 45 patients (ages 8-20 years), including 1 patient with stage II NPC and 44 patients with stage III/IV NPC, were recruited to the study. The patient with stage II disease received radiotherapy (59.4 grays [Gy]). The patients with stage III/IV disease received 3 courses of neoadjuvant chemotherapy with cisplatin, 5-fluorouracil, and folinic acid. The cumulative irradiation dose was 54 Gy in 5 patients, who achieved complete remission after neoadjuvant chemotherapy, and 59.4 Gy in the remaining 40 patients. All patients received concomitant cisplatin during the first week and last week of irradiation. After irradiation, all patients received interferon beta for 6 months. Tumor response was evaluated by magnetic resonance imaging studies and positron emission tomography scans.Results: After the completion of treatment, 43 of 45 patients were in complete remission. In 2 patients, only a partial response was achieved, followed by distant metastases (1 patient) or local progression and distant metastases (1 patient), 6 months and 10 months after diagnosis, respectively. Another patient developed a solitary pelvic bone metastasis 21 months after diagnosis. After a median follow-up of 30 months (range, 6-95 months), the event-free survival rate was 92.4%, and the overall survival was 97.1%. Acute toxicity consisted mainly of leucopenia, mucositis, and nausea; and late toxicity consisted of hearing loss and hypothyroidism.Conclusions: Combined therapy with neoadjuvant chemotherapy, radiochemotherapy, and interferon beta was well tolerated and resulted in a very good outcome that was superior to the outcomes of published results from all other pediatric NPC study groups. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
25. KCNQ1OT1 hypomethylation: a novel disguised genetic predisposition in sporadic pediatric adrenocortical tumors?
- Author
-
Wijnen M, Alders M, Zwaan CM, Wagner A, van den Heuvel-Eibrink MM, Wijnen, Mark, Alders, Mariëlle, Zwaan, Christian M, Wagner, Anja, and van den Heuvel-Eibrink, Marry M
- Published
- 2012
- Full Text
- View/download PDF
26. No Prognostic Impact of the WT1 Gene Single Nucleotide Polymorphism rs16754 in Pediatric Acute Myeloid Leukemia.
- Author
-
Hollink IH, van den Heuvel-Eibrink MM, Zimmermann M, Balgobind BV, Arentsen-Peters ST, Alders M, Willasch A, Kaspers GJ, Trka J, Baruchel A, Creutzig U, Pieters R, Reinhardt D, and Zwaan CM
- Published
- 2010
- Full Text
- View/download PDF
27. Low frequency of MLL-partial tandem duplications in paediatric acute myeloid leukaemia using MLPA as a novel DNA screenings technique.
- Author
-
Balgobind BV, Hollink IHI, Reinhardt D, van Wering ER, de Graaf SSN, Baruchel A, Stary J, Beverloo HB, de Greef GE, Pieters R, Zwaan CM, and van den Heuvel-Eibrink MM
- Abstract
Mixed-lineage leukaemia (MLL)-partial tandem duplications (PTDs) are found in 3-5% of adult acute myeloid leukaemia (AML), and are associated with poor prognosis. In adult AML, MLL-PTD is only detected in patients with trisomy 11 or internal tandem duplications of FLT3 (FLT3-ITD). To date, studies in paediatric AML are scarce, and reported large differences in the frequency of MLL-PTD, frequently utilising mRNA RT-PCR only to detect MLL-PTDs. We studied the frequency of MLL-PTD in a large cohort of paediatric AML (n=276) and the results from two different methods, i.e. mRNA RT-PCR, and multiplex ligation-dependent probe amplification (MLPA), a method designed to detect copy number differences of specific DNA sequences. In some patients with an MLL-rearrangement, MLL-PTD transcripts were detected, but were not confirmed by DNA-MLPA, indicating that DNA-MLPA can more accurately detect MLL-PTD compared to mRNA RT-PCR. In paediatric AML, MLL-PTD was detected in 7/276 patients (2.5%). One case had a trisomy 11, while the others had normal cytogenetics. Furthermore 4 of the 7 patients revealed a FLT3-ITD, which was significantly higher compared with the other AML cases (p=0.016). In conclusion, using DNA-MLPA as a novel screenings technique in combination with mRNA RT-PCR a low frequency of MLL-PTD in paediatric AML was found. Larger prospective studies are needed to further define the prognostic relevance of MLL-PTD in paediatric AML. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
28. t(6;9)(p22;q34)/DEK-NUP214 rearranged pediatric myeloid leukemia: an international study on 62 patients
- Author
-
Jean Michel Cayuela, Daisuke Tomizawa, Jochen Harbott, Der Cherng Liang, Christine Perot, Valerie de Haas, Nils von Neuhoff, Anne Auvrignon, Hiroto Inaba, Susana C. Raimondi, C. Michel Zwaan, Gertjan J.L. Kaspers, Christine J. Harrison, Gitte Kerndrup, Franco Locatelli, Marco Zecca, Souichi Adachi, Julie Damgaard Sandahl, Erik Forestier, Riccardo Masetti, Katarina Reinhardt, Lucy Chilton, Bertil Johansson, Maarten Fornerod, Marry M. van den Heuvel-Eibrink, Henrik Hasle, Eva A. Coenen, H. Berna Beverloo, Sandahl JD, Coenen EA, Forestier E, Harbott J, Johansson B, Kerndrup G, Adachi S, Auvrignon A, Beverloo HB, Cayuela JM, Chilton L, Fornerod M, de Haas V, Harrison CJ, Inaba H, Kaspers GJ, Liang DC, Locatelli F, Masetti R, Perot C, Raimondi S, Reinhardt K, Tomizawa D, von Neuhoff N, Zecca M, Zwaan CM, van den Heuvel-Eibrink MM, Hasle H, Erasmus MC other, Clinical Genetics, Pediatrics, Pediatric surgery, and CCA - Innovative therapy
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Myeloid ,Adolescent ,Oncogene Proteins, Fusion ,Chromosomal Proteins, Non-Histone ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Translocation, Genetic ,Bone Marrow ,Recurrence ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Cumulative incidence ,Child ,Poly-ADP-Ribose Binding Proteins ,Oncogene Proteins ,Hematology ,business.industry ,Myelodysplastic syndromes ,Gene Expression Profiling ,Childhood Acute Myeloid Leukemia ,Hematopoietic Stem Cell Transplantation ,Infant, Newborn ,Myeloid leukemia ,Infant ,Articles ,ACUTE NONLYMPHOCYTIC LEUKEMIA, TUMOR-SUPPRESSOR, GROWTH ARREST, PROTEIN DEK, FG REPEATS, PR DOMAIN, GENE, EXPRESSION, MALIGNANCIES, FUSION ,medicine.disease ,Nuclear Pore Complex Proteins ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,N/A ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,Leukemia, Myeloid ,Child, Preschool ,Myelodysplastic Syndromes ,Immunology ,Chromosomes, Human, Pair 6 ,Female ,business ,Chromosomes, Human, Pair 9 - Abstract
Acute myeloid leukemia with t(6;9)(p22;q34) is listed as a distinct entity in the 2008 World Health Organization classification, but little is known about the clinical implications of t(6;9)-positive myeloid leukemia in children. This international multicenter study presents the clinical and genetic characteristics of 62 pediatric patients with t(6;9)/DEK-NUP214-rearranged myeloid leukemia; 54 diagnosed as having acute myeloid leukemia, representing
- Published
- 2014
29. Menin inhibitors in pediatric acute leukemia: a comprehensive review and recommendations to accelerate progress in collaboration with adult leukemia and the international community.
- Author
-
Cuglievan B, Kantarjian H, Rubnitz JE, Cooper TM, Zwaan CM, Pollard JA, DiNardo CD, Kadia TM, Guest E, Short NJ, McCall D, Daver N, Nunez C, Haddad FG, Garcia M, Bhalla KN, Maiti A, Catueno S, Fiskus W, Carter BZ, Gibson A, Roth M, Khazal S, Tewari P, Abbas HA, Bourgeois W, Andreeff M, Shukla NN, Truong DD, Connors J, Ludwig JA, Stutterheim J, Salzer E, Juul-Dam KL, Sasaki K, Mahadeo KM, Tasian SK, Borthakur G, Dickson S, Jain N, Jabbour E, Meshinchi S, Garcia-Manero G, Ravandi F, Stein EM, Kolb EA, and Issa GC
- Subjects
- Humans, Child, Adult, Leukemia drug therapy, Leukemia genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Nucleophosmin
- Abstract
Aberrant expression of HOX and MEIS1 family genes, as seen in KMT2A-rearranged, NUP98-rearranged, or NPM1-mutated leukemias leads to arrested differentiation and leukemia development. HOX family genes are essential gatekeepers of physiologic hematopoiesis, and their expression is regulated by the interaction between KMT2A and menin. Menin inhibitors block this interaction, downregulate the abnormal expression of MEIS1 and other transcription factors and thereby release the differentiation block. Menin inhibitors show significant clinical efficacy against KMT2A-rearranged and NPM1-mutated acute leukemias, with promising potential to address unmet needs in various pediatric leukemia subtypes. In this collaborative initiative, pediatric and adult hematologists/oncologists, and stem cell transplant physicians have united their expertise to explore the potential of menin inhibitors in pediatric leukemia treatment internationally. Our efforts aim to provide a comprehensive clinical overview of menin inhibitors, integrating preclinical evidence and insights from ongoing global clinical trials. Additionally, we propose future international, inclusive, and efficient clinical trial designs, integrating pediatric populations in adult trials, to ensure broad access to this promising therapy for all children and adolescents with menin-dependent leukemias., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
30. Inotuzumab ozogamicin combined with chemotherapy in pediatric B-cell precursor CD22 + acute lymphoblastic leukemia: results of the phase IB ITCC-059 trial.
- Author
-
Pennesi E, Brivio E, Ammerlaan ACJ, Jiang Y, Van der Velden VHJ, Beverloo HB, Sleight B, Locatelli F, Brethon B, Rossig C, Engstler G, Nilsson A, Bruno B, Petit A, Bielorai B, Rizzari C, Rialland F, Rubio-San-Simón A, Sirvent FJB, Diaz-de-Heredia C, Rives S, and Zwaan CM
- Subjects
- Humans, Child, Male, Female, Child, Preschool, Adolescent, Treatment Outcome, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Vincristine administration & dosage, Vincristine therapeutic use, Infant, Inotuzumab Ozogamicin administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Sialic Acid Binding Ig-like Lectin 2 antagonists & inhibitors
- Abstract
Inotuzumab ozogamicin (InO) is a CD22-directed antibody conjugated with calicheamicin. The phase IB of the ITCC-059 trial tested InO combined with chemotherapy in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Relapsed /refractory CD22+ BCP-ALL pediatric patients were enrolled. The primary objective was to establish the recommended phase II dose (RP2D). Secondary objectives included preliminary efficacy and tolerability. InO was combined with 1.5 mg/m2 of vincristine (days 3, 10, 17, 24), 20 mg/m2 of dexamethasone (2 5-day blocks, then amended), and intrathecal therapy. A rolling-6 design was used testing InO from 0.8 to 1.8 mg/m2/cycle. Between May 2020 and April 2022, 30 patients were treated, and 29 were evaluable for dose limiting toxicities (DLT). At 1.1 mg/m2/cycle, two of four patients had DLT (liver toxicity). InO was de-escalated to 0.8 mg/m2/cycle (N=6) without DLT while awaiting a protocol amendment to reduce dexamethasone dose to 10 mg/m2. Post amendment, InO was re-escalated to 1.1 mg/m2/cycle (N=6, 1 DLT), then to 1.4 mg/m2/ cycle (N=3, no DLT), and finally to 1.8 mg/m2/cycle (N=7, 1 DLT). Three additional patients were treated in an expansion cohort. The pooled response rate was 80% (24/30; 95% confidence interval [CI]: 61.4-92.3) and, among responders, 66.7% achieved minimal residual disease negativity. The RP2D of InO combined with vincristine, dexamethasone and intrathecal therapy was declared at 1.8 mg/m2/cycle (1.5 mg/m2/cycle after remission) in a fractioned schedule. This combination showed a response rate similar to the single agent cohorts of this trial, with liver toxicity issues at the initial higher dexamethasone dose (clinicaltrials gov. Identifier: NTR5736).
- Published
- 2024
- Full Text
- View/download PDF
31. NUP98 oncofusions in myeloid malignancies: An update on molecular mechanisms and therapeutic opportunities.
- Author
-
Rasouli M, Troester S, Grebien F, Goemans BF, Zwaan CM, and Heidenreich O
- Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy with a heterogeneous molecular landscape. In the pediatric context, the NUP98 gene is a frequent target of chromosomal rearrangements that are linked to poor prognosis and unfavorable treatment outcomes in different AML subtypes. The translocations fuse NUP98 to a diverse array of partner genes, resulting in fusion proteins with novel functions. NUP98 fusion oncoproteins induce aberrant biomolecular condensation, abnormal gene expression programs, and re-wired protein interactions which ultimately cause alterations in the cell cycle and changes in cellular structures, all of which contribute to leukemia development. The extent of these effects is steered by the functional domains of the fusion partners and the influence of concomitant somatic mutations. In this review, we discuss the complex characteristics of NUP98 fusion proteins and potential novel therapeutic approaches for NUP98 fusion-driven AML., Competing Interests: Olaf Heidenreich received research funding from Syndax and Roche. The other authors declare no conflicts of interest., (© 2024 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2024
- Full Text
- View/download PDF
32. Precision cancer medicine platform trials: Concepts and design of AcSé-ESMART.
- Author
-
Geoerger B, Bautista F, André N, Berlanga P, Gatz SA, Marshall LV, Rubino J, Archambaud B, Marchais A, Rubio-San-Simón A, Ducassou S, Zwaan CM, Casanova M, Nysom K, Pellegrino S, Hoog-Labouret N, Buzyn A, Blanc P, Paoletti X, and Vassal G
- Subjects
- Humans, Biomarkers, Tumor genetics, Molecular Targeted Therapy methods, Research Design standards, Clinical Trials, Phase II as Topic, Proof of Concept Study, Precision Medicine methods, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Precision cancer medicine brought the promise of improving outcomes for patients with cancer. High-throughput molecular profiling of tumors at treatment failure aims to direct a patient to a treatment matched to the tumor profile. In this way, improved outcome has been achieved in a small number of patients whose tumors exhibit unique targetable oncogenic drivers. Most cancers, however, contain multiple genetic alterations belonging to and of various hallmarks of cancer; for most of these alterations, there is limited knowledge on the level of evidence, their hierarchical roles in oncogenicity, and utility as biomarkers for response to targeted treatment(s). We developed a proof-of-concept trial that explores new treatment strategies in a molecularly-enriched tumor-agnostic, pediatric population. The evaluation of novel agents, including first-in-child molecules, alone or in combination, is guided by the available understanding of or hypotheses for the mechanisms of action of the diverse cancer events. Main objectives are: to determine 1) recommended phase 2 doses, 2) activity signals to provide the basis for disease specific development, and 3) to define new predictive biomarkers. Here we outline concepts, rationales and designs applied in the European AcSé-ESMART trial and highlight the feasibility but also complexity and challenges of such innovative platform trials., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: BG has had an advisory role for AstraZeneca and IDMC roles for trials sponsored by Roche and Novartis. SAG has had an advisory role for EMD Serono/MERCK KGaA and AMGEN and received research funding (institution) from AstraZeneca, GSK and BAYER. LVM has had Advisory Board Honoraria for Bayer, BMS, Day One Biopharmaceuticals, Eli Lilly, Illumina, Novartis and Tesaro. Paediatric Preceptorship/Chair & Speaker's Honoraria - Bayer. IDMC member for clinical trials sponsored by Eisai and Merck. NA has had an advisory role for BAYER and Partners Therapeutics and receives grants (institution) from Bristol Myers Squibb and drugs for a trial from Bristol Myers Squibb, Pierre Fabre, Merck, Pfizer and travel support from Roche; he further has IDMC roles for Accord Healthcare. AR had a consulting role for EusaPharma, Sanofi and SERB. She received honoraria from EusaPharma and Roche for educational events and travel expenses. MC has had advisory roles for Astra Zeneca, Bayer, Pfizer and was invited speaker for Bayer. KN serves on a data monitoring committee for Lilly. All remaining authors have declared no conflict of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
33. A multidimensional analysis reveals distinct immune phenotypes and the composition of immune aggregates in pediatric acute myeloid leukemia.
- Author
-
Koedijk JB, van der Werf I, Penter L, Vermeulen MA, Barneh F, Perzolli A, Meesters-Ensing JI, Metselaar DS, Margaritis T, Fiocco M, de Groot-Kruseman HA, Moeniralam R, Bang Christensen K, Porter B, Pfaff K, Garcia JS, Rodig SJ, Wu CJ, Hasle H, Nierkens S, Belderbos ME, Zwaan CM, and Heidenreich O
- Abstract
Because of the low mutational burden and consequently, fewer potential neoantigens, children with acute myeloid leukemia (AML) are thought to have a T cell-depleted or 'cold' tumor microenvironment and may have a low likelihood of response to T cell-directed immunotherapies. Understanding the composition, phenotype, and spatial organization of T cells and other microenvironmental populations in the pediatric AML bone marrow (BM) is essential for informing future immunotherapeutic trials about targetable immune-evasion mechanisms specific to pediatric AML. Here, we conducted a multidimensional analysis of the tumor immune microenvironment in pediatric AML and non-leukemic controls. We demonstrated that nearly one-third of pediatric AML cases has an immune-infiltrated BM, which is characterized by a decreased ratio of M2- to M1-like macrophages. Furthermore, we detected the presence of large T cell networks, both with and without colocalizing B cells, in the BM and dissected the cellular composition of T- and B cell-rich aggregates using spatial transcriptomics. These analyses revealed that these aggregates are hotspots of CD8
+ T cells, memory B cells, plasma cells and/or plasmablasts, and M1-like macrophages. Collectively, our study provides a multidimensional characterization of the BM immune microenvironment in pediatric AML and indicates starting points for further investigations into immunomodulatory mechanisms in this devastating disease., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
34. Menin Inhibition With Revumenib for KMT2A -Rearranged Relapsed or Refractory Acute Leukemia (AUGMENT-101).
- Author
-
Issa GC, Aldoss I, Thirman MJ, DiPersio J, Arellano M, Blachly JS, Mannis GN, Perl A, Dickens DS, McMahon CM, Traer E, Zwaan CM, Grove CS, Stone R, Shami PJ, Mantzaris I, Greenwood M, Shukla N, Cuglievan B, Kovacsovics T, Gu Y, Bagley RG, Madigan K, Chudnovsky Y, Nguyen HV, McNeer N, and Stein EM
- Abstract
Purpose: Revumenib, an oral, small molecule inhibitor of the menin-lysine methyltransferase 2A (KMT2A) interaction, showed promising efficacy and safety in a phase I study of heavily pretreated patients with KMT2A -rearranged ( KMT2Ar ) acute leukemia. Here, we evaluated the activity of revumenib in individuals with relapsed/refractory (R/R) KMT2Ar acute leukemia., Methods: AUGMENT-101 is a phase I/II, open-label, dose-escalation and expansion study of revumenib conducted across 22 clinical sites in five countries (ClinicalTrials.gov identifier: NCT04065399). We report results from the phase II, registration-enabling portion. Individuals age ≥30 days with R/R KMT2Ar acute leukemia or with AML and nucleophosmin 1 ( NPM1 ) mutation were enrolled. Revumenib was administered once every 12 hours, at 163 mg (95 mg/m
2 if weight <40 kg) with a strong cytochrome P450 inhibitor, in 28-day cycles. The primary end points were the rate of complete remission (CR) or CR with partial hematologic recovery (CR + CRh) and safety. At a prespecified interim analysis, safety was assessed in all KMT2Ar treated patients; efficacy was assessed in those with centrally confirmed KMT2Ar . The separate NPM1 cohort of the trial is ongoing., Results: From October 1, 2021, to July 24, 2023, N = 94 patients (median [range] age, 37 [1.3-75] years) were treated. Grade ≥3 adverse events included febrile neutropenia (37.2%), differentiation syndrome (16.0%), and QTc prolongation (13.8%). In the efficacy-evaluable patients (n = 57), the CR + CRh rate was 22.8% (95% CI, 12.7 to 35.8), exceeding the null hypothesis of 10% ( P = .0036). Overall response rate was 63.2% (95% CI, 49.3 to 75.6), with 15 of 22 patients (68.2%) having no detectable residual disease., Conclusion: Revumenib led to high remission rates with a predictable safety profile in R/R KMT2Ar acute leukemia. To our knowledge, this trial represents the largest evaluation of a targeted therapy for these patients.- Published
- 2024
- Full Text
- View/download PDF
35. Immunoglobulin prophylaxis prevents hospital admissions for fever in pediatric acute lymphoblastic leukemia: results of a multicenter randomized trial.
- Author
-
Thus KA, De Groot-Kruseman HA, Winkler-Seinstra P, Fiocco M, Segers H, Van den Bos C, Van der Sluis IM, Tissing WJE, Veening MA, Zwaan CM, Van Tilburg CM, Pieters R, and Bierings M
- Abstract
Infections lead to substantial morbidity during treatment of acute lymphoblastic leukemia (ALL) in which the adaptive immune system gets severely affected, leading to declining serum immunoglobulin levels. The aim of this trial was to investigate whether intravenous immunoglobulin (IVIG) prophylaxis in pediatric patients with ALL prevents admissions for fever. This randomized controlled trial was a subtrial of the national Dutch multicenter ALL study. Patients aged 1-19 years with medium risk (MR) ALL were randomized into two groups receiving either IVIG prophylaxis (0.7 g/kg IVIG given every three weeks, starting day 22 after diagnosis) or well defined standard of care (control group). Between October 2012 until March 2019, 91 (51%) patients were randomly assigned to IVIG prophylaxis and 86 (49%) to the control arm. In the IVIG prophylaxis group there were 206 admissions for fever versus 271 in the control group (p=0.011). IVIG prophylaxis was not associated with bacteremia. However, IVIG prophylaxis was associated with significantly less admissions for fever with negative blood cultures compared to the control group (N=113 versus 200, p.
- Published
- 2024
- Full Text
- View/download PDF
36. Integrated drug resistance and leukemic stemness gene-expression scores predict outcomes in large cohort of over 3500 AML patients from 10 trials.
- Author
-
H Elsayed A, Cao X, Marrero RJ, Nguyen NHK, Wu H, Ni Y, Ribeiro RC, Tobias H, Valk PJ, Béliveau F, Richard-Carpentier G, Hébert J, Zwaan CM, Gamis A, Kolb EA, Aplenc R, Alonzo TA, Meshinchi S, Rubnitz J, Pounds S, and Lamba JK
- Abstract
In this study, we leveraged machine-learning tools by evaluating expression of genes of pharmacological relevance to standard-AML chemotherapy (ara-C/daunorubicin/etoposide) in a discovery-cohort of pediatric AML patients (N = 163; NCT00136084 ) and defined a 5-gene-drug resistance score (ADE-RS5) that was predictive of outcome (high MRD1 positivity p = 0.013; lower EFS p < 0.0001 and OS p < 0.0001). ADE-RS5 was integrated with a previously defined leukemic-stemness signature (pLSC6) to classify patients into four groups. ADE-RS5, pLSC6 and integrated-score was evaluated for association with outcome in one of the largest assembly of ~3600 AML patients from 10 independent cohorts (1861 pediatric and 1773 adult AML). Patients with high ADE-RS5 had poor outcome in validation cohorts and the previously reported pLSC6 maintained strong significant association in all validation cohorts. For pLSC6/ADE-RS5-integrated-score analysis, using Group-1 (low-scores for ADE-RS5 and pLSC6) as reference, Group-4 (high-scores for ADE-RS5 and pLSC6) showed worst outcome (EFS: p < 0.0001 and OS: p < 0.0001). Groups-2/3 (one high and one low-score) showed intermediate outcome (p < 0.001). Integrated score groups remained an independent predictor of outcome in multivariable-analysis after adjusting for established prognostic factors (EFS: Group 2 vs. 1, HR = 4.68, p < 0.001, Group 3 vs. 1, HR = 3.22, p = 0.01, and Group 4 vs. 1, HR = 7.26, p < 0.001). These results highlight the significant prognostic value of transcriptomics-based scores capturing disease aggressiveness through pLSC6 and drug resistance via ADE-RS5. The pLSC6 stemness score is a significant predictor of outcome and associates with high-risk group features, the ADE-RS5 drug resistance score adds further value, reflecting the clinical utility of simultaneous testing of both for optimizing treatment strategies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
37. Evolution of the Innovative Therapies for Children With Cancer Consortium Trial Portfolio for Drug Development for Children With Cancer.
- Author
-
Bautista F, Verdú-Amorós J, Geoerger B, Rubio-San-Simón A, Paoletti X, Zwaan CM, Casanova M, Marshall LV, Carceller F, Doz F, Lecinse C, Vassal G, Pearson ADJ, Kearns P, and Moreno L
- Subjects
- Humans, Child, Adolescent, Clinical Trials, Phase II as Topic, Antineoplastic Agents therapeutic use, Clinical Trials, Phase I as Topic, Therapies, Investigational, Research Design, Neoplasms drug therapy, Drug Development
- Abstract
Purpose: The aim of the Innovative Therapies for Children with Cancer (ITCC) consortium is to improve access to novel therapies for children and adolescents with cancer. The evolution of the ITCC clinical trial portfolio since 2003 was reviewed., Methods: All ITCC-labeled phase I/II trials opened between January 1, 2003 and February 3, 2018 were analyzed in two periods (2003-2010 and 2011-2018), and data were extracted from the ITCC database, regulatory agencies' registries, and publications., Results: Sixty-one trials (62% industry-sponsored) enrolled 3,198 patients. The number of trials in the second period increased by almost 300% (16 v 45). All biomarker-driven trials (n = 14) were conducted in the second period. The use of rolling six and model-based designs increased (1 of 9, 11% v 21 of 31, 68%), and that of 3 + 3 designs decreased (5 of 9, 55% v 5 of 31, 16%; P = .014). The proportion of studies evaluating chemotherapeutics only decreased (5 of 16, 31% v 4 of 45, 9%), the proportion of single-agent targeted therapies did not change (9 of 16, 56.2% v 24 of 45, 53.3%), the proportion of combination targeted therapies trials increased (2 of 16, 12%, v 17 of 45, 38%), the proportion of randomized phase II trials increased (1 of 7, 14% v 8 of 14, 57%). More trials were part of a pediatric investigation plan in the second period (4 of 16, 25% v 21 of 45, 46%). The median time for Ethics Committees' approvals was 1.7 times longer for academic compared with industry-sponsored trials., Conclusion: This study reports a shift in the paradigm of early drug development for childhood cancers, with more biologically relevant targets evaluated in biomarker-driven trials or in combination with other therapies and with more model-based or randomized designs and a greater focus on fulfilling regulatory requirements. Improvement of trial setup and recruitment could increase the number of patients benefiting from novel agents.
- Published
- 2024
- Full Text
- View/download PDF
38. Selective pressures of platinum compounds shape the evolution of therapy-related myeloid neoplasms.
- Author
-
Bertrums EJM, de Kanter JK, Derks LLM, Verheul M, Trabut L, van Roosmalen MJ, Hasle H, Antoniou E, Reinhardt D, Dworzak MN, Mühlegger N, van den Heuvel-Eibrink MM, Zwaan CM, Goemans BF, and van Boxtel R
- Subjects
- Humans, Child, Male, Female, Platinum Compounds therapeutic use, Adult, Adolescent, Whole Genome Sequencing, Phylogeny, Child, Preschool, Antineoplastic Agents therapeutic use, Single-Cell Analysis, Tumor Suppressor Protein p53 genetics, Li-Fraumeni Syndrome genetics, Germ-Line Mutation, Neoplasms, Second Primary genetics
- Abstract
Therapy-related myeloid neoplasms (t-MN) arise as a complication of chemo- and/or radiotherapy. Although t-MN can occur both in adult and childhood cancer survivors, the mechanisms driving therapy-related leukemogenesis likely vary across different ages. Chemotherapy is thought to induce driver mutations in children, whereas in adults pre-existing mutant clones are selected by the exposure. However, selective pressures induced by chemotherapy early in life are less well studied. Here, we use single-cell whole genome sequencing and phylogenetic inference to show that the founding cell of t-MN in children starts expanding after cessation of platinum exposure. In patients with Li-Fraumeni syndrome, characterized by a germline TP53 mutation, we find that the t-MN already expands during treatment, suggesting that platinum-induced growth inhibition is TP53-dependent. Our results demonstrate that germline aberrations can interact with treatment exposures in inducing t-MN, which is important for the development of more targeted, patient-specific treatment regimens and follow-up., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
39. Oncogenic and immunological targets for matched therapy of pediatric blood cancer patients: Dutch iTHER study experience.
- Author
-
Boer JM, Ilan U, Boeree A, Langenberg KPS, Koster J, Koudijs MJ, Hehir-Kwa JY, Nierkens S, Rossi C, Molenaar JJ, Goemans BF, den Boer ML, and Zwaan CM
- Abstract
Over the past 10 years, institutional and national molecular tumor boards have been implemented for relapsed or refractory pediatric cancer to prioritize targeted drugs for individualized treatment based on actionable oncogenic lesions, including the Dutch iTHER platform. Hematological malignancies form a minority in precision medicine studies. Here, we report on 56 iTHER leukemia/lymphoma patients for which we considered cell surface markers and oncogenic aberrations as actionable events, supplemented with ex vivo drug sensitivity for six patients. Prior to iTHER registration, 34% of the patients had received allogeneic hematopoietic cell transplantation (HCT) and 18% CAR-T therapy. For 51 patients (91%), a sample with sufficient tumor percentage (≥20%) required for comprehensive diagnostic testing was obtained. Up to 10 oncogenic actionable events were prioritized in 49/51 patients, and immunotherapy targets were identified in all profiled patients. Targeted treatment(s) based on the iTHER advice was given to 24 of 51 patients (47%), including immunotherapy in 17 patients, a targeted drug matching an oncogenic aberration in 12 patients, and a drug based on ex vivo drug sensitivity in one patient, resulting in objective responses and a bridge to HCT in the majority of the patients. In conclusion, comprehensive profiling of relapsed/refractory hematological malignancies showed multiple oncogenic and immunotherapy targets for a precision medicine approach, which requires multidisciplinary expertise to prioritize the best treatment options for this rare, heavily pretreated pediatric population., Competing Interests: C. Michel Zwaan has received research funding from Syndax, Abbvie, Takeda, Jazz Pharmaceutical, and Pfizer. C. Michel Zwaan has been involved as a consultant for BMS, Gilead, Kura Oncology, Novartis, Incyte, and Pfizer; C. Michel Zwaan has performed an advisory role for Sanofi and Novartis. C. Michel Zwaan is a member of the board of directors of the ITCC Hem Malignancies Committee. The other authors declare no conflicts of interest., (© 2024 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2024
- Full Text
- View/download PDF
40. When InO says no: understanding escape.
- Author
-
Boer JM and Zwaan CM
- Published
- 2024
- Full Text
- View/download PDF
41. Population Pharmacokinetics of Inotuzumab Ozogamicin in Pediatric Relapsed/Refractory B-Cell Precursor Acute Lymphoblastic Leukemia: Results of Study ITCC-059.
- Author
-
Wu JH, Pennesi E, Bautista F, Garrett M, Fukuhara K, Brivio E, Ammerlaan ACJ, Locatelli F, van der Sluis IM, Rossig C, Chen-Santel C, Bielorai B, Petit A, Starý J, Díaz-de-Heredia C, Rives S, O'Marcaigh A, Rizzari C, Engstler G, Nysom K, Rubio-San-Simón A, Bruno B, Bertrand Y, Brethon B, Rialland F, Plat G, Dirksen U, Sramkova L, Zwaan CM, and Huitema ADR
- Subjects
- Humans, Child, Male, Female, Adolescent, Child, Preschool, Adult, Young Adult, Middle Aged, Models, Biological, Recurrence, Infant, Aged, Inotuzumab Ozogamicin pharmacokinetics, Inotuzumab Ozogamicin administration & dosage, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma blood, Antineoplastic Agents, Immunological pharmacokinetics, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological therapeutic use
- Abstract
Background and Objective: Inotuzumab ozogamicin is an antibody-drug conjugate approved for treating relapsed/refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL) in adults. Pediatric pharmacokinetic data of inotuzumab ozogamicin are lacking. This study is the first to examine the population pharmacokinetics of inotuzumab ozogamicin in pediatric patients with relapsed/refractory BCP-ALL., Methods: From 531 adult patients with B-cell non-Hodgkin's lymphoma, 234 adult patients with BCP-ALL, and 53 pediatric patients with BCP-ALL, 8924 inotuzumab ozogamicin serum concentrations were analyzed using non-linear mixed-effects modeling. A published adult inotuzumab ozogamicin population-pharmacokinetic model, a two-compartment model with linear and time-dependent clearance, was adapted to describe the pediatric data., Results: Modifications in this analysis, compared to the published adult model, included: (i) re-estimating pharmacokinetic parameters and covariate effects; (ii) modifying covariate representation; and (iii) introducing relevant pediatric covariate effects (age on the decay coefficient of time-dependent clearance and ALL effect (disease type and/or different bioanalytical analysis methods) on initial values of time-dependent clearance). For patients with relapsed/refractory BCP-ALL, increasing age was associated with a decreasing decay coefficient of time-dependent clearance, reflecting that the target-mediated drug clearance declines more rapidly in children. In pediatric BCP-ALL, the median [interquartile range] cumulative area under the concentration-time curve was significantly higher among responders (n = 42) versus non-responders (n = 10) at the end of the first cycle (26.1 [18.9-35.0] vs 10.1 [9.19-16.1], × 10
3 ng*h/mL, p < 0.001). From simulations performed at the recommended pediatric phase II dose, inotuzumab ozogamicin exposure reached a similar level as observed in responding pediatric trial participants., Conclusions: The pharmacokinetic profile of inotuzumab ozogamicin in pediatric patients with relapsed/refractory BCP-ALL was well described in this study. No dose adjustment is required clinically for pediatric patients with BCP-ALL based on the simulated inotuzumab ozogamicin exposure at the recommended pediatric phase II dose, promising efficacy and acceptable tolerability., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
42. Optimized cytogenetic risk-group stratification of KMT2A-rearranged pediatric acute myeloid leukemia.
- Author
-
van Weelderen RE, Harrison CJ, Klein K, Jiang Y, Abrahamsson J, Alonzo T, Aplenc R, Arad-Cohen N, Bart-Delabesse E, Buldini B, De Moerloose B, Dworzak MN, Elitzur S, Fernández Navarro JM, Gamis A, Gerbing RB, Goemans BF, de Groot-Kruseman HA, Guest E, Ha SY, Hasle H, Kelaidi C, Lapillonne H, Leverger G, Locatelli F, Miyamura T, Norén-Nyström U, Polychronopoulou S, Rasche M, Rubnitz JE, Stary J, Tierens A, Tomizawa D, Zwaan CM, and Kaspers GJL
- Subjects
- Humans, Child, Male, Female, Child, Preschool, Adolescent, Infant, Prognosis, Chromosome Aberrations, Gene Rearrangement, Retrospective Studies, Myeloid-Lymphoid Leukemia Protein genetics, Histone-Lysine N-Methyltransferase genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality
- Abstract
Abstract: A comprehensive international consensus on the cytogenetic risk-group stratification of KMT2A-rearranged (KMT2A-r) pediatric acute myeloid leukemia (AML) is lacking. This retrospective (2005-2016) International Berlin-Frankfurt-Münster Study Group study on 1256 children with KMT2A-r AML aims to validate the prognostic value of established recurring KMT2A fusions and additional cytogenetic aberrations (ACAs) and to define additional, recurring KMT2A fusions and ACAs, evaluating their prognostic relevance. Compared with our previous study, 3 additional, recurring KMT2A-r groups were defined: Xq24/KMT2A::SEPT6, 1p32/KMT2A::EPS15, and 17q12/t(11;17)(q23;q12). Across 13 KMT2A-r groups, 5-year event-free survival probabilities varied significantly (21.8%-76.2%; P < .01). ACAs occurred in 46.8% of 1200 patients with complete karyotypes, correlating with inferior overall survival (56.8% vs 67.9%; P < .01). Multivariable analyses confirmed independent associations of 4q21/KMT2A::AFF1, 6q27/KMT2A::AFDN, 10p12/KMT2A::MLLT10, 10p11.2/KMT2A::ABI1, and 19p13.3/KMT2A::MLLT1 with adverse outcomes, but not those of 1q21/KMT2A::MLLT11 and trisomy 19 with favorable and adverse outcomes, respectively. Newly identified ACAs with independent adverse prognoses were monosomy 10, trisomies 1, 6, 16, and X, add(12p), and del(9q). Among patients with 9p22/KMT2A::MLLT3, the independent association of French-American-British-type M5 with favorable outcomes was confirmed, and those of trisomy 6 and measurable residual disease at end of induction with adverse outcomes were identified. We provide evidence to incorporate 5 adverse-risk KMT2A fusions into the cytogenetic risk-group stratification of KMT2A-r pediatric AML, to revise the favorable-risk classification of 1q21/KMT2A::MLLT11 to intermediate risk, and to refine the risk-stratification of 9p22/KMT2A::MLLT3 AML. Future studies should validate the associations between the newly identified ACAs and outcomes and unravel the underlying biological pathogenesis of KMT2A fusions and ACAs., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
43. Targeting the innate immune system in pediatric and adult AML.
- Author
-
Perzolli A, Koedijk JB, Zwaan CM, and Heidenreich O
- Subjects
- Humans, Adult, Child, Killer Cells, Natural immunology, Immunity, Innate, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute therapy, Immunotherapy methods, Tumor Microenvironment immunology
- Abstract
While the introduction of T cell-based immunotherapies has improved outcomes in many cancer types, the development of immunotherapies for both adult and pediatric AML has been relatively slow and limited. In addition to the need to identify suitable target antigens, a better understanding of the immunosuppressive tumor microenvironment is necessary for the design of novel immunotherapy approaches. To date, most immune characterization studies in AML have focused on T cells, while innate immune lineages such as monocytes, granulocytes and natural killer (NK) cells, received less attention. In solid cancers, studies have shown that innate immune cells, such as macrophages, myeloid-derived suppressor cells and neutrophils are highly plastic and may differentiate into immunosuppressive cells depending on signals received in their microenvironment, while NK cells appear to be functionally impaired. Hence, an in-depth characterization of the innate immune compartment in the TME is urgently needed to guide the development of immunotherapeutic interventions for AML. In this review, we summarize the current knowledge on the innate immune compartment in AML, and we discuss how targeting its components may enhance T cell-based- and other immunotherapeutic approaches., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
44. Naked antibodies and antibody-drug conjugates: targeted therapy for childhood acute lymphoblastic leukemia.
- Author
-
Brivio E, Bautista F, and Zwaan CM
- Subjects
- Humans, Child, Antineoplastic Agents, Immunological therapeutic use, Treatment Outcome, Inotuzumab Ozogamicin therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Molecular Targeted Therapy, Immunoconjugates therapeutic use, Antibodies, Bispecific therapeutic use
- Abstract
The treatment of childhood acute lymphoblastic leukemia (ALL) has reached overall survival rates exceeding 90%. The present and future challenges are to cure the remainder of patients still dying from disease, and to reduce morbidity and mortality in those who can be cured with standard-of-care chemotherapy by replacing toxic chemotherapy elements while retaining cure rates. With the novel therapeutic options introduced in the last years, including immunotherapies and targeted antibodies, the treatment of ALL is undergoing major changes. For B-cell precursor ALL, blinatumomab, an anti-CD19 bispecific antibody, has established its role in the consolidation treatment for both high- and standard-risk first relapse of ALL, in the presence of bone marrow involvement, and may also have an impact on the outcome of high-risk subsets such as infant ALL and Philadelphia chromosome-positive ALL. Inotuzumab ozogamicin, an anti-CD22 drug conjugated antibody, has demonstrated high efficacy in inducing complete remission in relapsed ALL, even in the presence of high tumor burden, but randomized phase III trials are still ongoing. For T-ALL the role of CD38-directed treatment, such as daratumumab, is gaining interest, but randomized data are needed to assess its specific benefit. These antibodies are currently being tested in patients with newly diagnosed ALL and may lead to major changes in the present paradigm of treatment of pediatric ALL. Unlike the past, lessons may be learned from innovations in adult ALL, in which more drastic changes are piloted that may need to be translated to pediatrics.
- Published
- 2024
- Full Text
- View/download PDF
45. Distinct Responses to Menin Inhibition and Synergy with DOT1L Inhibition in KMT2A -Rearranged Acute Lymphoblastic and Myeloid Leukemia.
- Author
-
Adriaanse FRS, Schneider P, Arentsen-Peters STCJM, Fonseca AMND, Stutterheim J, Pieters R, Zwaan CM, and Stam RW
- Subjects
- Humans, Drug Synergism, Gene Rearrangement, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Drug Resistance, Neoplasm drug effects, Mutation, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Methyltransferases antagonists & inhibitors, Methyltransferases genetics, Methyltransferases metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology
- Abstract
Pediatric acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) exhibit favorable survival rates. However, for AML and ALL patients carrying KMT2A gene translocations clinical outcome remains unsatisfactory. Key players in KMT2A-fusion-driven leukemogenesis include menin and DOT1L. Recently, menin inhibitors like revumenib have garnered attention for their potential therapeutic efficacy in treating KMT2A -rearranged acute leukemias. However, resistance to menin inhibition poses challenges, and identifying which patients would benefit from revumenib treatment is crucial. Here, we investigated the in vitro response to revumenib in KMT2A -rearranged ALL and AML. While ALL samples show rapid, dose-dependent induction of leukemic cell death, AML responses are much slower and promote myeloid differentiation. Furthermore, we reveal that acquired resistance to revumenib in KMT2A -rearranged ALL cells can occur either through the acquisition of MEN1 mutations or independently of mutations in MEN1 . Finally, we demonstrate significant synergy between revumenib and the DOT1L inhibitor pinometostat in KMT2A -rearranged ALL, suggesting that such drug combinations represent a potent therapeutic strategy for these patients. Collectively, our findings underscore the complexity of resistance mechanisms and advocate for precise patient stratification to optimize the use of menin inhibitors in KMT2A -rearranged acute leukemia.
- Published
- 2024
- Full Text
- View/download PDF
46. Efficacy and safety of azacitidine in pediatric patients with newly diagnosed advanced myelodysplastic syndromes before hematopoietic stem cell transplantation in the AZA-JMML-001 trial.
- Author
-
Locatelli F, Strålin KB, Schmid I, Sevilla J, Smith OP, van den Heuvel-Eibrink MM, Zecca M, Zwaan CM, Gaudy A, Patturajan M, Poon J, Simcock M, and Niemeyer CM
- Subjects
- Humans, Child, Azacitidine adverse effects, Treatment Outcome, Leukemia, Myeloid, Acute drug therapy, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes diagnosis, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Here we report efficacy, pharmacokinetics, and safety data obtained in treatment-naive, pediatric patients with newly diagnosed advanced MDS receiving azacitidine in the AZA-JMML-001 study. The primary endpoint was response rate (proportion of patients with complete response [CR], partial response [PR], or marrow CR, sustained for ≥4 weeks). Of the 10 patients enrolled, one had an unconfirmed marrow CR and none had confirmed responses after three cycles; the study was therefore closed after stage 1. Azacitidine was well tolerated. The lack of efficacy of azacitidine in pediatric patients with newly diagnosed advanced MDS highlights the need for effective new treatments in these patients., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
47. A 14-year-old girl with premature ovarian insufficiency but with a positive pregnancy test.
- Author
-
Touwslager RNH, Zwaan CM, Bakker B, Lentjes EGWM, Looijenga LHJ, and van Santen HM
- Subjects
- Humans, Female, Adolescent, Pregnancy, Pregnancy Tests, Neuroblastoma complications, Neuroblastoma pathology, Neuroblastoma drug therapy, Adrenal Gland Neoplasms complications, Adrenal Gland Neoplasms pathology, Adrenal Gland Neoplasms diagnosis, False Positive Reactions, Luteinizing Hormone blood, Prognosis, Primary Ovarian Insufficiency diagnosis, Primary Ovarian Insufficiency pathology
- Abstract
Objectives: Childhood cancer survivors are at risk for premature ovarian insufficiency, especially after treatment with alkylating agents. The objective of this report is to highlight a case in which this phenomenon caused a false-positive pregnancy test., Case Presentation: A workup was performed in a 14-year-old girl with a positive pregnancy test. She was diagnosed with stage IV neuroblastoma of the left adrenal gland at the age of 4 years. She received extensive treatment, including alkylating agents, and had been diagnosed with premature ovarian insufficiency. An LH/hCG suppression test was performed using high dose 17 bèta-estradiol: hCG levels normalized., Conclusions: The pregnancy test was false-positive due to production of low amounts of hCG by the pituitary gland as a result of high LH concentrations following premature ovarian insufficiency. It may be helpful to perform the LH/hCG suppression test to prove pituitary origin of the hCG overproduction., (© 2024 the author(s), published by De Gruyter, Berlin/Boston.)
- Published
- 2024
- Full Text
- View/download PDF
48. Bevacizumab, Irinotecan, or Topotecan Added to Temozolomide for Children With Relapsed and Refractory Neuroblastoma: Results of the ITCC-SIOPEN BEACON-Neuroblastoma Trial.
- Author
-
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, and Wheatley K
- Subjects
- Child, Humans, Infant, Child, Preschool, Adolescent, Young Adult, Adult, Temozolomide therapeutic use, Irinotecan therapeutic use, Bevacizumab adverse effects, Dacarbazine adverse effects, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Topotecan adverse effects, Neuroblastoma pathology
- 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.
- Published
- 2024
- Full Text
- View/download PDF
49. Bosutinib in Resistant and Intolerant Pediatric Patients With Chronic Phase Chronic Myeloid Leukemia: Results From the Phase I Part of Study ITCC054/COG AAML1921.
- Author
-
Brivio E, Pennesi E, Willemse ME, Huitema ADR, Jiang Y, van Tinteren HDR, van der Velden VHJ, Beverloo BH, den Boer ML, Rammeloo LAJ, Hudson C, Heerema N, Kowalski K, Zhao H, Kuttschreuter L, Bautista Sirvent FJ, Bukowinski A, Rizzari C, Pollard J, Murillo-Sanjuán L, Kutny M, Zarnegar-Lumley S, Redell M, Cooper S, Bertrand Y, Petit A, Krystal J, Metzler M, Lancaster D, Bourquin JP, Motwani J, van der Sluis IM, Locatelli F, Roth ME, Hijiya N, and Zwaan CM
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Humans, Infant, Aniline Compounds adverse effects, Nitriles adverse effects, Protein Kinase Inhibitors adverse effects, Treatment Outcome, Antineoplastic Agents adverse effects, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myeloid, Chronic-Phase drug therapy, Quinolines adverse effects
- Abstract
Purpose: Bosutinib is approved for adults with chronic myeloid leukemia (CML): 400 mg once daily in newly diagnosed (ND); 500 mg once daily in resistant/intolerant (R/I) patients. Bosutinib has a different tolerability profile than other tyrosine kinase inhibitors (TKIs) and potentially less impact on growth (preclinical data). The primary objective of this first-in-child trial was to determine the recommended phase II dose (RP2D) for pediatric R/I and ND patients., Patients and Methods: In the phase I part of this international, open-label trial (ClinicalTrials.gov identifier: NCT04258943), children age 1-18 years with R/I (per European LeukemiaNet 2013) Ph+ CML were enrolled using a 6 + 4 design, testing 300, 350, and 400 mg/m
2 once daily with food. The RP2D was the dose resulting in 0/6 or 1/10 dose-limiting toxicities (DLTs) during the first cycle and achieving adult target AUC levels for the respective indication. As ND participants were only enrolled in phase II, the ND RP2D was selected based on data from R/I patients., Results: Thirty patients were enrolled; 27 were evaluable for DLT: six at 300 mg/m2 , 11 at 350 mg/m2 (one DLT), and 10 at 400 mg/m2 (one DLT). The mean AUCs at 300 mg/m2 , 350 mg/m2 , and 400 mg/m2 were 2.20 μg h/mL, 2.52 μg h/mL, and 2.66 μg h/mL, respectively. The most common adverse event was diarrhea (93%; ≥grade 3: 11%). Seven patients stopped because of intolerance and eight because of insufficient response. Complete cytogenetic and major molecular response to bosutinib appeared comparable with other published phase I/II trials with second-generation TKIs in children., Conclusion: Bosutinib was safe and effective. The pediatric RP2D was 400 mg/m2 once daily (max 600 mg/d) with food in R/I patients and 300 mg/m2 once daily (max 500 mg/d) with food in ND patients, which achieved targeted exposures as per adult experience.- Published
- 2024
- Full Text
- View/download PDF
50. Acute myeloid leukemias with UBTF tandem duplications are sensitive to menin inhibitors.
- Author
-
Barajas JM, Rasouli M, Umeda M, Hiltenbrand R, Abdelhamed S, Mohnani R, Arthur B, Westover T, Thomas ME 3rd, Ashtiani M, Janke LJ, Xu B, Chang TC, Rosikiewicz W, Xiong E, Rolle C, Low J, Krishan R, Song G, Walsh MP, Ma J, Rubnitz JE, Iacobucci I, Chen T, Krippner-Heidenreich A, Zwaan CM, Heidenreich O, and Klco JM
- Subjects
- Humans, Child, Transcription Factors, Myeloid Ecotropic Viral Integration Site 1 Protein genetics, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology
- Abstract
Abstract: UBTF tandem duplications (UBTF-TDs) have recently emerged as a recurrent alteration in pediatric and adult acute myeloid leukemia (AML). UBTF-TD leukemias are characterized by a poor response to conventional chemotherapy and a transcriptional signature that mirrors NUP98-rearranged and NPM1-mutant AMLs, including HOX-gene dysregulation. However, the mechanism by which UBTF-TD drives leukemogenesis remains unknown. In this study, we investigated the genomic occupancy of UBTF-TD in transformed cord blood CD34+ cells and patient-derived xenograft models. We found that UBTF-TD protein maintained genomic occupancy at ribosomal DNA loci while also occupying genomic targets commonly dysregulated in UBTF-TD myeloid malignancies, such as the HOXA/HOXB gene clusters and MEIS1. These data suggest that UBTF-TD is a gain-of-function alteration that results in mislocalization to genomic loci dysregulated in UBTF-TD leukemias. UBTF-TD also co-occupies key genomic loci with KMT2A and menin, which are known to be key partners involved in HOX-dysregulated leukemias. Using a protein degradation system, we showed that stemness, proliferation, and transcriptional signatures are dependent on sustained UBTF-TD localization to chromatin. Finally, we demonstrate that primary cells from UBTF-TD leukemias are sensitive to the menin inhibitor SNDX-5613, resulting in markedly reduced in vitro and in vivo tumor growth, myeloid differentiation, and abrogation of the UBTF-TD leukemic expression signature. These findings provide a viable therapeutic strategy for patients with this high-risk AML subtype., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.