PURPOSE
There is an increasing need to evaluate innovative drugs for childhood cancer using combination strategies. Strong biological rationale and clinical experience suggest that multiple agents will be more efficacious than monotherapy for most diseases and may overcome resistance mechanisms and increase synergy. The process to evaluate these combination trials needs to maximize efficiency and should be agreed by all stakeholders.
METHODS
After a review of existing combination trial methodologies, regulatory requirements, and current results, a consensus among stakeholders was achieved.
RESULTS
Combinations of anticancer therapies should be developed on the basis of mechanism of action and robust preclinical evaluation, and may include data from adult clinical trials. The general principle for combination early-phase studies is that, when possible, clinical trials should be dose- and schedule-confirmatory rather than dose-exploratory, and every effort should be made to optimize doses early. Efficient early-phase combination trials should be seamless, including dose confirmation and randomized expansion. Dose evaluation designs for combinations depend on the extent of previous knowledge. If not previously evaluated, limited evaluation of monotherapy should be included in the same clinical trial as the combination. Randomized evaluation of a new agent plus standard therapy versus standard therapy is the most effective approach to isolate the effect and toxicity of the novel agent. Platform trials may be valuable in the evaluation of combination studies. Patient advocates and regulators should be engaged with investigators early in a proposed clinical development pathway and trial design must consider regulatory requirements.
CONCLUSION
An optimized, agreed approach to the design and evaluation of early-phase pediatric combination trials will accelerate drug development and benefit all stakeholders, most importantly children and adolescents with cancer.
INTRODUCTION
There is an urgent need to develop new therapies for children and adolescents with cancer, both to improve outcome for poor prognosis malignancies1 and to reduce acute and long-term adverse effects of current treatments.2,3 Biological rationale and published experience indicate that combination approaches, particularly those that use agents with robust single-agent activity,4–6 will be more efficacious than the same agents used alone. Therefore, there is an increasing need to evaluate innovative drugs for childhood cancer using combinatorial strategies. The adult drug development landscape has radically evolved over the past decade, moving away from a traditional approach to seamless-design, phase I/II trials for single agents.7 Seamless trials can address multiple objectives under the heading of early-phase investigation and allow for rapid completion.
CONTEXT
Key Objective
To achieve an international consensus among stakeholders on the process to design and deliver early-phase combination trials in children and adolescents with cancer.
Knowledge Generated
An efficient, effective approach on the basis of mechanism of action and robust preclinical evaluation is recommended to evaluate new combinations of anticancer agents and identify the efficacy and toxicity of each novel agent. The very early involvement of patient advocates and regulators in a proposed clinical development pathway and trial design is crucial.
Relevance (S. Bhatia)
-
This paper provides general guidance to accelerate the availability of optimized treatments for children and adolescents with cancer.*
*Relevance section written by JCO Associate Editor Smita Bhatia, MD, MPH, FASCO.
Consensus articles have described the conduct of early-phase trials in children to bring beneficial innovative agents more rapidly to the clinic.8,9 With the increasing evaluation of agents in combination, there is a need for a consensus to combination early-phase trials. New combinations may be developed with existing standard-of-care chemotherapy regimens or with other new agents (novel-novel combinations). The goal should be to evaluate drug combinations rapidly and efficiently, without compromising identification of safety signals or assuming unnecessary risks of toxicity. Ideally, this should require the smallest number of patients to enroll to determine safe dose(s) and schedule, identify early signals of activity, and reach a go/no-go decision in a tumor or target-specific population.
In addition to efficient trial designs, three changes are needed to accelerate evaluation of innovative medicines in children. The first change requires that the development of anticancer medicines and combinations for children be based on the biology of childhood cancers and the mechanism of action of the drug(s) investigated rather than the adult indication.10,11 The second requires that new drugs with high potential for benefit be assessed quickly and early in the context of the development in adults. A third improvement is expanding in vivo preclinical testing using genomically characterized pediatric models to provide the strongest possible data set for prioritizing specific combinations among the multitude of potential combinations that could be evaluated clinically.12 These changes can reduce the unacceptable delay (median 6.5 years) from the initiation of first-in-human trials to first-in-child trials.13 Legislation in Europe,14,15 and more recently in the United States,16 prioritizes science-driven, patient-oriented, pediatric oncology drug development over adult-indication approaches. These regulatory advances, together with the international multistakeholder organization ACCELERATE, promote children having greater access to innovative, safe, and effective treatments.17,18
Early engagement of regulators in the clinical development of agents for pediatric cancers is critical. Trial design needs to consider regulatory requirements (pediatric investigation plan and initial pediatric study plans) along a full clinical development pathway including early- and late-phase combination trials for novel agents, and for all drugs included in the combination. By aligning scientific, regulatory, and payer (eg, European health technology assessment bodies) requirements from the inception of a clinical trial, the fewest number of patients will need to be enrolled to obtain sufficient evidence for scientific and regulatory purposes.18 Similarly, involving patient advocates early and throughout discussions is important for both pragmatic and principled reasons.19 Doing so ensures the patient voice is heard and that specific unmet needs are accounted for in all phases of pediatric oncology drug development.18
Herein, we report a consensus opinion among invested stakeholders and discuss salient points that should be considered in the development of early-phase combination trials for pediatric oncology, presenting best practices whenever possible.
SUMMARY OF RECOMMENDED PRACTICE FOR GENERAL PEDIATRIC ONCOLOGY EARLY-PHASE TRIALS
Prior position statements have strongly advocated that the pediatric clinical evaluation of a new anticancer drug follows an early-phase seamless trial design rather than individual phase I and II trials.10,20–22 An early-phase clinical trial has two components: (1) a dose-finding or dose-confirmation phase and (2) expansion cohort(s) (Table 1) in which optimal dosing, toxicity profile, pharmacokinetic (PK) parameters, pharmacodynamic (PD) effects, and early signals of antitumor activity are obtained. The therapeutic agent is then transitioned to late-stage trials to determine antitumor efficacy and comparison with current standard(s) of care. Avoiding the distinction between different phases of development can reduce the numbers of exposed patients and as well limit cost, resource requirements, trial development timelines, and trial duration. Monotherapy or combination platform trials23 with several parallel arms24–28 are examples of potential frameworks with many advantages (Table 2).
TABLE 1.
General Principles of Pediatric Early-Phase Clinical Trials
TABLE 2.
Advantages of Platform Trials for Clinical Drug Development in Children With Cancer

In practice, no differences have been observed in the PK of cytotoxic drugs between patients age 5-11 and 11-16 years, or between 12-16 and 16-21 years.29–33 Furthermore, the median age of enrollment was 12 years or younger in 15 of 20 recent Innovative Therapies for Children with Cancer (ITCC)–published phase I trials (F. Bautista, personal communication, January 2023). Therefore, to expedite dose-finding for pediatric patients, age-specific cohorts for the majority of pediatric patients are discouraged. Additional PK/PD data can be collected in infants age 2 years and younger, who are anticipated to have different metabolism, using dedicated PK/PD expansion cohorts at the recommended phase II dose (RP2D) and in later-phase studies.
Early-phase trials should also avoid defining a lower age limit for inclusion, unless biologically justified. To this end, developing age-appropriate formulations is critical. However, this development should not delay the start of the first-in-child trial. Rather, trials should begin using tablets or capsules, feasible to reliably deliver a pediatric dose with adult formulations. When a child-friendly formulation becomes available with bioequivalence data, this should be used and PK data for the new formulation collected subsequently.
A mechanism of action, tumor-agnostic development approach provides more opportunities for rapid and focused pediatric and adult drug development, if the relevant biomarker is also age-agnostic.34,35
CONSIDERATIONS FOR PEDIATRIC COMBINATION TRIALS
General Principles
Early-phase combination trials, like monotherapy trials, should be seamless and encompass both a dose-finding/confirmation and an activity evaluation phase. Different approaches are required for safely combining agents on the basis of prior clinical experience with the agent or agents-in-class. Combinations may contain agents previously studied in children as monotherapy, or those for which there are only adult data. When agents are combined, new toxicities can occur that are not present when agents are administered individually, and overlapping toxicity is possible. For combinations of a novel drug with standard-of-care chemotherapy, scenarios depend on whether the pediatric PK and safety profile of the new drugs is known, and if overlapping toxicities or PK interactions are expected. Novel-novel combinations are indicated when there is a strong biological rationale with robust preclinical data and are particularly compelling if early proof-of-principle clinical data exist.36 For novel-novel combinations, scenarios depend on whether the products have (1) known pediatric PK and safety profiles; (2) known adult PK and safety profiles; (3) metabolism that is expected to contribute to PK interactions; and (4) observed or expected interactions or overlapping toxicities (Table 3, Fig 1).
TABLE 3.
Clinical Scenarios and Proposed Design and Starting Dose Strategies
FIG 1.
Spectrum of designs and starting dose for pediatric combination trials. The design depends on the amount of known existing data relating to the drug and the target/entity in pediatrics and in adults, expectations for interactions or overlapping toxicities, and the number of drugs for which dose and schedule will be explored. At one end, there are second- or third-in-class products where there are pediatric data on the class and the emphasis is on dose confirmation. In the middle of the spectrum, only one drug is escalated for which there is some information from trials in adults, but some potential safety concerns or expected drug interactions—here, dose finding is required but could potentially start at the adult recommended dose for the same combination with planned escalation or de-escalation using an approach such as a rolling six design if the number of dose levels is smaller (only 2-3.) At the other end of the spectrum, there are combinations of novel-novel agents studied where there is no experience in adults and more than one drug is to be escalated or de-escalated; in this case, a partial ordering CRM approach is appropriate starting at a lower dose than adult equivalent RP2D. CRM, continual reassessment method; RP2D, recommended phase II dose.
Parents enroll their children on early-phase clinical trials in the hope that participation will directly benefit them.37,38 Although the goal of a trial is to address a scientific question, therapeutic intent (ie, the potential for patient benefit, balanced against potential or real short- and long-term toxicity) should guide trial design considerations. This means conducting appropriate patient selection during dose-finding to maximize chances of benefit, avoiding subtherapeutic doses, minimizing single-agent evaluations, except when necessary, and rapidly assessing agent efficacy. To limit the number of patients who receive a dose less than a potentially beneficial treatment, intrasubject dose escalation should be considered. Maximum dose escalation may also not be required if the dose used in adults (if any) provides evidence of biological activity and is supported by PK. A unique aspect of combination trials is to demonstrate signals of improved activity over either monotherapy or standard of care. Randomization is of value and follows the European Medicine Agency (EMA) guidelines for combination development.36 Therapeutic intent is also enhanced in the context of combination studies of existing plus novel drugs if there is benefit with existing drug(s) alone. Randomized evaluation of a new agent plus standard therapy versus standard therapy is the most effective approach to isolate the effect and toxicity of the novel agent.36 Early-phase platform studies,24–28 where arms of different drugs can be evaluated, provide an efficient way for combinations to be evaluated for efficacy in relatively small patient populations and dropped rapidly if there is no early signal. Continual dialogue is required to challenge the perception that a phase I study has no realistic potential for benefit.37,38 Early-phase testing must be designed with an eye toward where the combination might ultimately fit into existing treatment paradigms for newly diagnosed patients.10
As with all pediatric clinical trials, incorporating translational correlative research is a crucial element of early-phase trials, so that knowledge can be increased to guide future evaluation of more biologically rational combination regimens. Moreover, this approach will facilitate the detailed retrospective molecular analyses of responders and nonresponders to generate a revised or new hypothesis.
Rationale for Combinations
Selection of combinations with compelling biological and clinical rationale for evaluation in children is essential, given the rarity of pediatric cancers and the mismatch between the immense numbers of combinations that are available for testing compared with the number of clinical trials that can be conducted. Prioritization of agents should be based on knowledge of tumor biology, molecular drivers of disease, a drug's mechanism of action, robust activity of the combination in relevant in vivo preclinical models that exceeds that of the component agents used alone, and therapeutic unmet needs.10 Another important factor is the single-agent activity of the drugs used in the combination, as it is uncommon for meaningful clinical benefit to be observed for combinations in which an agent lacking evidence of single-agent activity is evaluated.4,39,40 Recent advances from positive pediatric phase III trials highlight the importance of the activity of the agent added to standard-of-care regimens, as illustrated by rituximab for non-Hodgkin lymphoma,41 brentuximab vedotin for high-risk Hodgkin lymphoma,42 arsenic trioxide for acute promyelocytic leukemia,43 blinatumomab for B-cell acute lymphoblastic leukemia (ALL),44,45 nelarabine for T-ALL,46 and imatinib47/disatinib48 for Philadelphia chromosome–positive ALL and dinutuximab with chemotherapy for neuroblastoma.49
Evidence generated nonclinically is of paramount importance with the objective that the activity of the combination is at least additive or synergistic. Although synergy observed in preclinical studies may highlight combinations of potential interest, it is important to consider that the synergy observed for cancer cell lines in vitro may also apply to one or more critical normal tissues when combinations are tested in patients and translate into toxicity, with the result that there is little or no therapeutic window for the combinations. For this reason, in vivo studies with appropriate controls are required. An example is O6-benzylguanine plus nitrosoureas for high-grade gliomas. Remarkable preclinical synergy was observed for O6-benzylguanine plus nitrosourea combinations,50 but in patients, the synergistic effect also applied to normal hematopoietic cells leading to excessive myelosuppression that required reduced doses of nitrosourea when biologically relevant doses of O6-benzylguanine were administered.51 A phase III trial comparing O6-benzylguanine plus reduced-dose bischloronitrosourea, carmustine (BCNU) to full-dose BCNU for adults with high-grade glioma was stopped early for futility.52 Other examples of preclinical synergy with excessive clinical toxicity are the combination of CHK1 inhibitors with gemcitabine,53 MEK inhibitors with pan-HER inhibitors,54–57 imatinib/dasatinib with high-dose chemotherapy,58 and crizotinib with vinblastine in anaplastic large-cell lymphoma.59
The extent and depth of the preclinical studies required for support of combinations depend on the strength of the underlying biological hypothesis. For example, if the target of an agent that is being incorporated into a combination is a well-defined oncogenic driver, then less evidence may be required. Conversely, in the majority of instances, when the agent lacks a predictive biomarker of response, robust preclinical evidence for a substantial combination effect is warranted before moving forward to clinical testing. Prioritization should be given to combinations with evidence of synthetic lethality, where inhibiting two targets together results in cell death, but inhibiting one target alone does not.60 The ITCC-P4 and Pediatric Preclinical Testing Consortium12,61,62 consensus on minimum preclinical testing requirements for the development of investigational therapies can facilitate combination strategies.
Design Considerations
Inclusion criteria.
Depending on preclinical and adult trial data, the pediatric dose-confirmation/finding phase may include an enriched selected population24,63 or all-comers.64 Another approach is to include all-comers in the dose-confirmation/finding phase and an enriched population for histology-/genomic-specific expansions to determine activity.65,66 If there is biological rationale and potential benefit for the individual patient, such as non–biomarker-driven agents, all-comers may facilitate accrual and quickly ascertain a safe and adequate drug exposure dosing schedule. If preclinical or adult data suggest that antitumor activity is highly unlikely in an unselected population, eligibility criteria should be restricted to the relevant target population.63 Biomarker-positive or histology-specific patients may be permitted to enter the study at any time at the best current estimate of a safe dose when there is no available slot in the main open cohort.67,68 To maximally use these data flexibly, statistical methods such as model-guided methods69 (continual reassessment method [CRM]-type) may be used. When determining antitumor activity, the population should be restricted by disease or by biomarker to the target population for future clinical development or there should be partial enrichment to enroll a proportion of patients whose tumor has the target of interest. Patients who have prior exposure to single-agent therapy may be permitted, as this may reveal the benefit of the combination, by demonstrating response in a previously resistant population.70
Defining the starting dose.
Confirmation of the adult body surface area (BSA)– or weight-adjusted RP2D should be the preferred strategy rather than dose finding. More conservative dose finding, starting at 20%-30% lower than the adult RP2D, may be warranted if there are safety concerns, a narrow therapeutic index, overlapping toxicities, or expected drug-drug interactions. The dose-confirmation approach will enable shorter studies.71–73
The sample size of the dose-confirmation cohort should enable the estimation of PK parameters and the determination of the recommended dose for the combination (RDC) for children.
Dose and schedule finding.
In line with EMA guidelines,36 studies should aim to identify the product(s) causing the observed adverse reactions to guide dose reductions in relation to observed toxicity. For example, if one agent is particularly likely to cause an observed adverse event (eg, rash), it should be de-escalated first. Generally, the novel product should be dose escalated/de-escalated first when a novel agent is combined with a known active agent or backbone. In addition, preclinical evidence of mechanism-based synergistic toxicity should be considered, for example, that seen with talazoparib and temozolomide.65 Designs to de-escalate each component of the combination regimen if there is toxicity, or escalate if the exposure is less than occurring in adults or inadequate target inhibition, need to be pre-specified, so that trials are not halted unnecessarily.31,74
The concept of an acceptable toxicity should be considered, independently of its grade as per Common Terminology Criteria for Adverse Events. For example, reversible hematologic toxicity could be acceptable, in contrast to a permanent cardiac toxicity, which would be unacceptable. Class effects should be considered; for example, dose-limiting toxicity (DLT) definitions should exclude known class-related side effects easily manageable with supportive care. The concept of tolerability is also important, but potentially subjective, and might be measured as a quality-of-life metric. Increasing input from advocates and use of patient-reported outcomes is also encouraged, particularly for symptom-based toxicities for which concordance is poor between patient and clinician or caregiver reports.75–77
Preclinical evaluations for additivity and synergy can guide optimal exposures for combinations. However, optimal combination dose and schedule finding in patients may require simultaneous exploration of dose for each drug and multiple schedules. A CRM design for unknown toxicity ordering (partial ordering) is efficient in exploring dose combinations with as few patients as possible.78 Dose-escalation strategies may include either cohorts that use alternate increases of each drug (or changes in schedule) or use of one drug in its standard single-agent dose and schedule and increase only the dose of the novel drug(s). Generally, if there are only two or three dose levels, then the rolling six design79 can be acceptable. However, if there are more dose levels for the combination, a partial ordering CRM (POCRM) approach78 (Table 3) is typically more accurate in identifying the combination dose and schedule and allows for tailoring which drug is escalated or de-escalated at completion of each cohort.80 The POCRM can also be designed to avoid waiting lists, testing multiple doses and schedules in parallel.
RDC.
It is not always possible to establish an optimal biological dose (eg, dose generating an adequate PD response without excessive toxicity) on the basis of PD data.81 Despite a strong biological rationale and convincing preclinical work, generating exposure/PD response data in the target population may not be feasible if there are no relevant/validated biomarkers. Preclinical data and response data from clinical trials in adults may provide information to determine the RDC in pediatrics. Dose optimization and dose-finding rely on toxicity, tolerability,36 and interpatient variability in exposure to target adult PK exposure with therapeutic drug monitoring whenever feasible.
Traditionally, the maximum tolerated dose (MTD) is defined on DLTs observed during cycle one of the dose-escalation phase, but the RDC should incorporate all information (eg, severe or chronic cumulative toxicity after cycle 1, dose modifications, PD and PK data, and toxicities that have significant impact on quality of life) since repeated dose reductions at cycles ≥2 are often because of chronic toxicity/intolerability that are not accounted in the definition of the MTD.82 Dose optimization might also enable intrapatient dose escalation (after achieving steady-state drug exposure or completion of two cycles and response evaluation) in patients who tolerate drugs and when therapeutic drug monitoring suggests a higher dose would be preferable.83,84
It is highly important to characterize doses and schedules of molecularly targeted therapy before initiating registration trials, as has been highlighted by Project Optimus of the US Food and Drug Administration (FDA).85 An inadequately characterized dose and schedule of one or more combination agents may lead to more toxicity without additional efficacy, resulting in unwanted consequences, for example, persistent or irreversible toxicities and inappropriate discontinuation of a potentially effective medicinal product.86 Given the size of clinical trials needed to reliably optimize dose and schedule, this optimization generally needs to occur in adult cancer populations with extrapolation of the optimal dosage- and exposure-response relationships for efficacy and toxicity to pediatric patients.22
Designs of trials with an initial monotherapy phase.
It is proposed that if a product is included in the combination where there are no previous pediatric data, a limited monotherapy evaluation, including an appropriate single-agent window, is included before proceeding to combination to better characterize dose, PK, toxicity, and, in some cases, single-agent activity. The starting dose for the monotherapy component should, in the absence of agent-specific toxicity, pharmacologic considerations, or a narrow therapeutic index, be 100% of the BSA-adjusted adult RP2D.9,21 Designs should allow for transition from monotherapy to combination therapy in the same patient as soon as possible (after one or two courses) unless substantial single-agent activity is observed. The need to determine efficacy of monotherapy will depend upon whether its preclinical mechanism of action is only to enhance the activity of other agents in the combination, or it is active on its own, with the caveat that there are few examples of successful development of agents without single-agent activity that are used only to enhance the activity of other agents. The minimum possible number of patients should receive monotherapy87,88 unless there is good evidence of single-agent activity and lack of evolving resistance to single-agent therapy.
Summary of approach for selection of starting dose and dose-finding designs.
Table 3 and Figure 1 illustrate potential scenarios for early-phase combination trials depending upon a range of agent characteristics.
An FDA guidance on the codevelopment of two or more new investigational drugs for use in combination89 is relevant to the developmental and regulatory pathways of novel-novel combinations in children, where there are no known pediatric data for at least one agent. This guidance also emphasizes the crucial importance of determining the contribution of each individual new investigational drug.
Evaluation of antitumor activity in early-phase combination trials.
Randomized evaluation of a new agent plus standard therapy versus standard therapy is most effective to isolate the effect of the addition of the novel agent in a combination.36,71 An underpowered randomization is considered acceptable and already agreed by the EMA.22,90 Crossover from standard to combination regimen after progression of disease may be allowed.91 This approach provides more robust evidence to identify promising regimens to take forward to later-stage trials and reduces the confounding effects of trial outcomes with unknown and uncontrollable trial effects such as patient selection, prior treatment, age, sex, comorbidities, referral bias, and differences in supportive care,72 compared with single-arm phase II trials. Relatively small, randomized expansion phases, randomized selection, or screening designs92,93 can be very valuable in screening for activity of a novel agent added to standard therapy. Novel designs (such as Bayesian or two-stage minimax Jung designs)94,95 can be used to minimize the sample size to 25-35 patients per cohort depending on the objectives and assumptions used in sample size determination. In this setting, controlling type I error (false positive) is less relevant than controlling type II error (false negative), as the goal of such trials is to ensure that if one regimen is superior, then there is a high probability that it will be selected. Pick-the-winner phase II randomized trial designs are another approach.96 Randomization of patients two to one to receive the arm combined with the innovative agent versus standard of care, respectively, could be considered to accelerate accrual if patients are more willing/inclined to enroll. However, this approach might lower the power or increase the total sample size to maintain the same power97 and should only be used if the activity of the investigational agent is expected to be high. The success or failure criteria generally are defined by clinically acceptable response rate or progression-free survival98 that would lead to further evaluation of the drug. Intercontinental studies are required to recruit sufficient numbers of patients in studies with low incidence.
Evaluating activity using Ensign (3-stage design allowing for two interim analyses implemented)99 or Simon's two-stage design100 via a single-arm trial using robust historical control or population known to be resistant to the single agent has several disadvantages, particularly in the absence of meaningful control data.
Patients in the dose-confirmation portion of the study can be included in the expansion cohort if they received the pediatric RDC and have disease status appropriate for assessment of the response endpoint. All responses and nonresponses (even those observed at lower dose level) should be reported, and prolonged disease stabilizations in some instances may be relevant.
Existing pediatric combination early-phase trials.
From a total of 287 trials including children in the ClinicalTrials.gov database, examples of published or presented pediatric combination early-phase trials are shown in Table 4.110
TABLE 4.
Selected Published or Presented Pediatric Combination Early-Phase Trials Reviewed to Identify Key Themes in Combination Trial Design
The phase I study of regorafenib in combination with vincristine and irinotecan was an amendment to single-agent regorafenib trial (ITCC-047). The combination used sequential dosing, had liberal DLT definitions,70 and moved rapidly to a second-line treatment in rhabdomyosarcoma (compared with standard of care) in a platform trial (FaR-RMS).111
The phase I/II study lenvatinib with etoposide plus ifosfamide (ITCC-050) included a combination dose-finding phase and a combination expansion in patients with osteosarcoma.101,102 This trial was followed by the OLIE (ITCC-082) randomized study evaluating the combination of lenvatinib with ifosfamide and etoposide compared with ifosfamide and etoposide alone (in relapsed/refractory osteosarcoma112). This efficient design could have been further accelerated if the randomized comparison had been integrated into the initial protocol and followed the phase I/II component.
The dabrafenib and trametinib combination trial began with a single-agent cohort to determine the RP2D of trametinib. This led to a limited dose escalation of dabrafenib and trametinib, followed by an expansion of the combination in patients with BRAF V600-mutant low-grade glioma.63 The combination was demonstrated to have a superior overall response rate and median progression-free survival when randomized against standard of care (carboplatin and vincristine) in pediatric low-grade gliomas.113
In conclusion, children and adolescents with cancer deserve early access to innovative drugs in clinical trials, and those agents that are potentially beneficial need to be evaluated expeditiously. Clinical trials should maximize the potential for benefit in the greatest possible proportion of patients contributing to such trials. Combinations should be developed on the basis of mechanism of action, cancer biology, robust preclinical evaluation, and clinical activity for the agent when known (Table 5). We propose that optimally efficient early-phase combination trials combine dose-confirmation/finding and randomized expansion cohorts in the tumor or target of interest.
TABLE 5.
Key Principles for Early-Phase Combination Trials in Pediatric Oncology
Very early discussion of trial designs with regulators is essential so that trials fulfill both scientific and regulatory purposes. Including parent advocates meaningfully in these early stages will help reveal potential points of confusion and misinformation, help shape strategies to increase recruitment and educate study participants, and help raise the likelihood of on-time completion of planned enrollment. Furthermore, this approach would allow children with rare conditions available to participate in clinical trials to be evaluated in the most parsimonious way possible.
A fit-for-purpose approach to the design of early-phase pediatric combination trials will benefit all stakeholders, especially children and adolescents with cancer.
ACKNOWLEDGMENT
The authors very gratefully acknowledge Gynette Cook for preparation of the manuscript.
Lucas Moreno
Honoraria: Bayer (Inst), EUSA Pharma (Inst)
Consulting or Advisory Role: Shionogi (Inst), Novartis (Inst), Boehringer Ingelheim (Inst), Norgine (Inst)
Patents, Royalties, Other Intellectual Property: Member of SIOPEN Executive Committee, SIOPEN receives royalties for the sales of dinutuximab beta (Inst)
Uncompensated Relationships: Ymabs Therapeutics Inc (Inst)
Steven G. DuBois
Consulting or Advisory Role: Bayer, Amgen, Jazz Pharmaceuticals
Research Funding: Merck (Inst), Roche/Genentech (Inst), Lilly (Inst), Curis (Inst), Loxo (Inst), BMS (Inst), Eisai (Inst), Pfizer (Inst), Turning Point Therapeutics (Inst), Bayer (Inst), Salarius Pharmaceuticals (Inst)
Travel, Accommodations, Expenses: Roche/Genentech, Salarius Pharmaceuticals
Uncompensated Relationships: Ymabs Therapeutics Inc
Julia Glade Bender
Consulting or Advisory Role: Jazz Pharmaceuticals
Research Funding: Eisai (Inst), Lilly (Inst), Loxo (Inst), Roche/Genentech (Inst), Bayer (Inst), Jazz Pharmaceuticals (Inst)
Patents, Royalties, Other Intellectual Property: Patent on a T lympboblastic lymphoma cell line, CUTLL1
Travel, Accommodations, Expenses: Amgen (Inst)
Uncompensated Relationships: SpringWorks Therapeutics, Bristol Myers Squibb, Merck, Eisai, Pfizer
Open Payments Link: https://openpaymentsdata.cms.gov/physician/708514
Audrey Mauguen
Research Funding: ImaginAb (Inst), Amgen (Inst)
Patents, Royalties, Other Intellectual Property: coinventor of provisional patent Number 63/193,700, filed on 5/27/21; conversion deadline: 5/27/22; named “Soothsayer,” filed by Office of Technology Development, MSKCC
Michela Casanova
Consulting or Advisory Role: Bayer, BMS, AstraZeneca, Servier, Pfizer
François Doz
Consulting or Advisory Role: ROCHE GENNENTECH (Inst), SERVIER (Inst), Ipsen (Inst), Novartis (Inst)
Research Funding: ONXEO (Inst)
Travel, Accommodations, Expenses: Bayer
Lia Gore
Stock and Other Ownership Interests: Amgen, Sanofi, Mirati Therapeutics, OnKure
Consulting or Advisory Role: Novartis, Amgen, Roche/Genentech, Syndax, OnKure, Janssen Oncology, Pfizer
Patents, Royalties, Other Intellectual Property: Patent held for diagnostic discovery and treatment response methodology tools in the use of MR spectroscopy for leukemia
Uncompensated Relationships: Kestrel Therapeutics
Douglas S. Hawkins
Research Funding: Bayer (Inst), Lilly (Inst), Jazz Pharmaceuticals (Inst), Pfizer (Inst)
Shai Izraeli
Honoraria: Foundation Medicine
Speakers' Bureau: Oncotest/Rhenium
David T.W. Jones
Patents, Royalties, Other Intellectual Property: Patent WO 2013075237 A1, titled “Mutations of histone proteins associated with proliferative disorders,” Patent WO2016142533A1, titled “DNA methylation-based method for classifying tumor species”
Pamela R. Kearns
Consulting or Advisory Role: Bristol Myers Squibb, Bayer, AstraZeneca
Research Funding: Pfizer (Inst), Bayer (Inst)
Karsten Nysom
Honoraria: Y-mAbs Therapeutics
Consulting or Advisory Role: Bayer, EUSA Pharma, Ymabs Therapeutics Inc, Lilly
Stefan Pfister
Stock and Other Ownership Interests: Heidelberg Epignostix
Consulting or Advisory Role: BioSkryb
Research Funding: Lilly (Inst), Bayer (Inst), Roche (Inst), PharmaMar (Inst), Pfizer (Inst), AstraZeneca (Inst), Janssen & Janssen (Inst), Servier (Inst), Sanofi (Inst), Amgen (Inst)
Patents, Royalties, Other Intellectual Property: patent on usng DNA methylation profiling for tumor classification, patent using nanopore sequencing for rapid tumor diagnostics
Gilles Vassal
Consulting or Advisory Role: Bayer, Roche/Genentech, AstraZeneca, Bristol Myers Squibb, Lilly, Novartis, Pfizer, Hutchinson-Medi Pharma, Pyramid
Travel, Accommodations, Expenses: Bristol Myers Squibb, Roche, Bayer
Christian Michel Zwaan
Consulting or Advisory Role: Takeda (Inst), Pfizer (Inst), AbbVie (Inst), Jazz Pharmaceuticals (Inst), Incyte (Inst), Novartis (Inst), Kura Oncology (Inst)
Research Funding: Takeda (Inst), AbbVie/Genentech (Inst), Pfizer (Inst), Jazz Pharmaceuticals (Inst), Kura Oncology (Inst), Daiichi Sankyo (Inst)
Xavier Paoletti
Honoraria: MSD Oncology
Consulting or Advisory Role: MSD Oncology, Daiichii Sankyo
Speakers' Bureau: Ipsen
Alexia Iasonos
This author is a deputy editor for Journal of Clinical Oncology. Journal policy recused the author from having any role in the peer review of this manuscript
Stock and Other Ownership Interests: Bristol Myers Squibb/Sanofi
Consulting or Advisory Role: Intelligencia, Mirati Therapeutics
Andrew D.J. Pearson
Honoraria: Lilly, Amgen, Developmental Therapeutics Consortium, Norgine
Consulting or Advisory Role: Lilly, Amgen, Developmental Therapeutics Consortium, Norgine
No other potential conflicts of interest were reported.
DISCLAIMER
The views expressed in this article are the personal views of the authors and may not be understood or quoted as being made on behalf of, or reflecting the position of, the agencies or organizations with which the authors are affiliated.
L.M., S.G.D., J.G.B., and A.M. are joint first authors.
AUTHOR CONTRIBUTIONS
Conception and design: Lucas Moreno, Steven G. DuBois, Julia Glade Bender, Audrey Mauguen, Xavier Paoletti, Alexia Iasonis, Andrew D.J. Pearson
Provision of study materials or patients: All authors
Data analysis and interpretation: All authors
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Combination Early-Phase Trials of Anticancer Agents in Children and Adolescents
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Lucas Moreno
Honoraria: Bayer (Inst), EUSA Pharma (Inst)
Consulting or Advisory Role: Shionogi (Inst), Novartis (Inst), Boehringer Ingelheim (Inst), Norgine (Inst)
Patents, Royalties, Other Intellectual Property: Member of SIOPEN Executive Committee, SIOPEN receives royalties for the sales of dinutuximab beta (Inst)
Uncompensated Relationships: Ymabs Therapeutics Inc (Inst)
Steven G. DuBois
Consulting or Advisory Role: Bayer, Amgen, Jazz Pharmaceuticals
Research Funding: Merck (Inst), Roche/Genentech (Inst), Lilly (Inst), Curis (Inst), Loxo (Inst), BMS (Inst), Eisai (Inst), Pfizer (Inst), Turning Point Therapeutics (Inst), Bayer (Inst), Salarius Pharmaceuticals (Inst)
Travel, Accommodations, Expenses: Roche/Genentech, Salarius Pharmaceuticals
Uncompensated Relationships: Ymabs Therapeutics Inc
Julia Glade Bender
Consulting or Advisory Role: Jazz Pharmaceuticals
Research Funding: Eisai (Inst), Lilly (Inst), Loxo (Inst), Roche/Genentech (Inst), Bayer (Inst), Jazz Pharmaceuticals (Inst)
Patents, Royalties, Other Intellectual Property: Patent on a T lympboblastic lymphoma cell line, CUTLL1
Travel, Accommodations, Expenses: Amgen (Inst)
Uncompensated Relationships: SpringWorks Therapeutics, Bristol Myers Squibb, Merck, Eisai, Pfizer
Open Payments Link: https://openpaymentsdata.cms.gov/physician/708514
Audrey Mauguen
Research Funding: ImaginAb (Inst), Amgen (Inst)
Patents, Royalties, Other Intellectual Property: coinventor of provisional patent Number 63/193,700, filed on 5/27/21; conversion deadline: 5/27/22; named “Soothsayer,” filed by Office of Technology Development, MSKCC
Michela Casanova
Consulting or Advisory Role: Bayer, BMS, AstraZeneca, Servier, Pfizer
François Doz
Consulting or Advisory Role: ROCHE GENNENTECH (Inst), SERVIER (Inst), Ipsen (Inst), Novartis (Inst)
Research Funding: ONXEO (Inst)
Travel, Accommodations, Expenses: Bayer
Lia Gore
Stock and Other Ownership Interests: Amgen, Sanofi, Mirati Therapeutics, OnKure
Consulting or Advisory Role: Novartis, Amgen, Roche/Genentech, Syndax, OnKure, Janssen Oncology, Pfizer
Patents, Royalties, Other Intellectual Property: Patent held for diagnostic discovery and treatment response methodology tools in the use of MR spectroscopy for leukemia
Uncompensated Relationships: Kestrel Therapeutics
Douglas S. Hawkins
Research Funding: Bayer (Inst), Lilly (Inst), Jazz Pharmaceuticals (Inst), Pfizer (Inst)
Shai Izraeli
Honoraria: Foundation Medicine
Speakers' Bureau: Oncotest/Rhenium
David T.W. Jones
Patents, Royalties, Other Intellectual Property: Patent WO 2013075237 A1, titled “Mutations of histone proteins associated with proliferative disorders,” Patent WO2016142533A1, titled “DNA methylation-based method for classifying tumor species”
Pamela R. Kearns
Consulting or Advisory Role: Bristol Myers Squibb, Bayer, AstraZeneca
Research Funding: Pfizer (Inst), Bayer (Inst)
Karsten Nysom
Honoraria: Y-mAbs Therapeutics
Consulting or Advisory Role: Bayer, EUSA Pharma, Ymabs Therapeutics Inc, Lilly
Stefan Pfister
Stock and Other Ownership Interests: Heidelberg Epignostix
Consulting or Advisory Role: BioSkryb
Research Funding: Lilly (Inst), Bayer (Inst), Roche (Inst), PharmaMar (Inst), Pfizer (Inst), AstraZeneca (Inst), Janssen & Janssen (Inst), Servier (Inst), Sanofi (Inst), Amgen (Inst)
Patents, Royalties, Other Intellectual Property: patent on usng DNA methylation profiling for tumor classification, patent using nanopore sequencing for rapid tumor diagnostics
Gilles Vassal
Consulting or Advisory Role: Bayer, Roche/Genentech, AstraZeneca, Bristol Myers Squibb, Lilly, Novartis, Pfizer, Hutchinson-Medi Pharma, Pyramid
Travel, Accommodations, Expenses: Bristol Myers Squibb, Roche, Bayer
Christian Michel Zwaan
Consulting or Advisory Role: Takeda (Inst), Pfizer (Inst), AbbVie (Inst), Jazz Pharmaceuticals (Inst), Incyte (Inst), Novartis (Inst), Kura Oncology (Inst)
Research Funding: Takeda (Inst), AbbVie/Genentech (Inst), Pfizer (Inst), Jazz Pharmaceuticals (Inst), Kura Oncology (Inst), Daiichi Sankyo (Inst)
Xavier Paoletti
Honoraria: MSD Oncology
Consulting or Advisory Role: MSD Oncology, Daiichii Sankyo
Speakers' Bureau: Ipsen
Alexia Iasonos
This author is a deputy editor for Journal of Clinical Oncology. Journal policy recused the author from having any role in the peer review of this manuscript
Stock and Other Ownership Interests: Bristol Myers Squibb/Sanofi
Consulting or Advisory Role: Intelligencia, Mirati Therapeutics
Andrew D.J. Pearson
Honoraria: Lilly, Amgen, Developmental Therapeutics Consortium, Norgine
Consulting or Advisory Role: Lilly, Amgen, Developmental Therapeutics Consortium, Norgine
No other potential conflicts of interest were reported.
REFERENCES
- 1.Noom AM, Howlader N, Krapcho M, et al. (eds): SEER Cancer Statistics Review, 1975-2015. Bethesda, MD, National Cancer Institute, 2018. https://seer.cancer.gov/csr/1975_2015/ [Google Scholar]
- 2.Williams AM, Liu Q, Bhakta N, et al. : Rethinking success in pediatric oncology: Beyond 5-year survival. J Clin Oncol 39:2227-2231, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Yeh JM, Ward ZJ, Chaudhry A, et al. : Life expectancy of adult survivors of childhood cancer over 3 decades. JAMA Oncol 6:350-357, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Foster J, Freidlin B, Korn EL, et al. : Evaluation of the contribution of randomised cancer clinical trials evaluating agents without documented single-agent activity. ESMO Open 5:e000871, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Plana D, Palmer AC, Sorger PK: Independent drug action in combination therapy: Implications for precision oncology. Cancer Discov 12:606-624, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Frei E, 3rd, Karon M, Levin RH, et al. : The effectiveness of combinations of antileukemic agents in inducing and maintaining remission in children with acute leukemia. Blood 26:642-656, 1965 [PubMed] [Google Scholar]
- 7.Prowell TM, Theoret MR, Pazdur R: Seamless oncology-drug development. N Engl J Med 374:2001-2003, 2016 [DOI] [PubMed] [Google Scholar]
- 8.Smith M, Bernstein M, Bleyer WA, et al. : Conduct of Phase I trials in children with cancer. J Clin Oncol 16:966-978, 1998 [DOI] [PubMed] [Google Scholar]
- 9.Moreno L, Pearson ADJ, Paoletti X, et al. : Early phase clinical trials of anticancer agents in children and adolescents—An ITCC perspective. Nat Rev Clin Oncol 14:497-507, 2017 [DOI] [PubMed] [Google Scholar]
- 10.Pearson AD, Herold R, Rousseau R, et al. : Implementation of mechanism of action biology-driven early drug development for children with cancer. Eur J Cancer 62:124-131, 2016 [DOI] [PubMed] [Google Scholar]
- 11.Pearson ADJ, Pfister SM, Baruchel A, et al. : From class waivers to precision medicine in paediatric oncology. Lancet Oncol 18:e394-e404, 2017 [DOI] [PubMed] [Google Scholar]
- 12.Vassal G, Houghton PJ, Pfister SM, et al. : International consensus on minimum preclinical testing requirements for the development of innovative therapies for children and adolescents with cancer. Mol Cancer Ther 20:1462F-1468F, 2021 [DOI] [PubMed] [Google Scholar]
- 13.Neel DV, Shulman DS, DuBois SG: Timing of first-in-child trials of FDA-approved oncology drugs. Eur J Cancer 112:49-56, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Regulation (EC) No 1901/2006 of the European Parliament and of the Council of 12 December 2006 on Medicinal Products for Paediatric Use and Amending Regulation (EEC) No 1768/92, Directive 2001/20/EC, Directive 2001/83/EC and Regulation (EC) No 726/2004 (Text with EEA relevance). https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A32006R1901&qid=1685131056256 [Google Scholar]
- 15.European Medicines Agency : European Medicines Agency decision (CW/0001/2015) of 23 July 2015 on class waivers, in accordance with Regulation (EC) No 1901/2006 of the European Parliament and of the Council. http://www.ema.europa.eu/docs/en_GB/document_library/Other/2015/07/WC500190385.pdf [Google Scholar]
- 16.US Congress: FDA Reauthorization Act of 2017. https://www.congress.gov/115/plaws/publ52/PLAW-115publ52.pdf
- 17.Vassal G, Rousseau R, Blanc P, et al. : Creating a unique, multi-stakeholder Paediatric Oncology Platform to improve drug development for children and adolescents with cancer. Eur J Cancer 51:218-224, 2015 [DOI] [PubMed] [Google Scholar]
- 18.Pearson ADJ, Weiner SL, Adamson PC, et al. : Accelerate—Five years accelerating cancer drug development for children and adolescents. Eur J Cancer 166:145-164, 2022 [DOI] [PubMed] [Google Scholar]
- 19.Deverka PA, Bangs R, Kreizenbeck K, et al. : A new framework for patient engagement in cancer clinical trials cooperative group studies. J Natl Cancer Inst 110:553-559, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rossoni C, Bardet A, Geoerger B, et al. : Sequential or combined designs for phase I/II clinical trials? A simulation study. Clin Trials 16:635-644, 2019 [DOI] [PubMed] [Google Scholar]
- 21.Paoletti X, Geoerger B, Doz F, et al. : A comparative analysis of paediatric dose-finding trials of molecularly targeted agent with adults’ trials. Eur J Cancer 49:2392-2402, 2013 [DOI] [PubMed] [Google Scholar]
- 22.European Medicines Agency : Reflection Paper on Extrapolation of Efficacy and Safety in Paediatric Medicine Development. https://www.ema.europa.eu/en/documents/scientific-guideline/draft-ich-guideline-e11a-pediatric-extrapolation-step-2b_en [Google Scholar]
- 23.The Adaptive Platform Trials Coalition : Adaptive platform trials: Definition, design, conduct and reporting considerations. Nat Rev Drug Discov 18:797-807, 2019 [DOI] [PubMed] [Google Scholar]
- 24.Bautista F, Paoletti X, Rubino J, 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 39:3546-3560, 2021 [DOI] [PubMed] [Google Scholar]
- 25.European proof-of-concept therapeutic stratification trial of molecular anomalies in relapsed or refractory tumors (ESMART)
- 26.Morscher RJ, Brard C, Berlanga P, 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 157:268-277, 2021 [DOI] [PubMed] [Google Scholar]
- 27.Parsons DW, Janeway KA, Patton DR, et al. : Actionable tumor alterations and treatment protocol enrollment of pediatric and young adult patients with refractory cancers in the national cancer institute-children's oncology group pediatric MATCH trial. J Clin Oncol 40:2224-2234, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Eckstein OS, Allen CE, Williams PM, et al. : Phase II study of selumetinib in children and young adults with tumors harboring activating mitogen-activated protein kinase pathway genetic alterations: Arm E of the NCI-COG pediatric MATCH trial. J Clin Oncol 40:2235-2245, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Doz F, Gentet JC, Pein F, et al. : Phase I trial and pharmacological study of a 3-hour paclitaxel infusion in children with refractory solid tumours: A SFOP study. Br J Cancer 84:604-610, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gore L, Trippett TM, Katzenstein HM, et al. : A multicenter, first-in-pediatrics, phase 1, pharmacokinetic and pharmacodynamic study of ridaforolimus in patients with refractory solid tumors. Clin Cancer Res 19:3649-3658, 2013 [DOI] [PubMed] [Google Scholar]
- 31.Mossé YP, Lim MS, Voss SD, et al. : Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: A children's oncology group phase 1 consortium study. Lancet Oncol 14:472-480, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mossé YP, Lipsitz E, Fox E, et al. : Pediatric phase I trial and pharmacokinetic study of MLN8237, an investigational oral selective small-molecule inhibitor of aurora kinase A: A children's oncology group phase I consortium study. Clin Cancer Res 18:6058-6064, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Vassal G, Doz F, Frappaz D, et al. : A phase I study of irinotecan as a 3-week schedule in children with refractory or recurrent solid tumors. J Clin Oncol 21:3844-3852, 2003 [DOI] [PubMed] [Google Scholar]
- 34.Pestana RC, Sen S, Hobbs BP, et al. : Histology-agnostic drug development—Considering issues beyond the tissue. Nat Rev Clin Oncol 17:555-568, 2020 [DOI] [PubMed] [Google Scholar]
- 35.Laetsch TW, DuBois SG, Mascarenhas L, et al. : Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: Phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol 19:705-714, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.EMA Anticancer Guideline (EMA/CHMP/205/95 Rev.6). https://www.ema.europa.eu/documents/scientific-guideline/draft-guideline-evaluation-anticancer-medicinal-products-man-revision-6_en.pdf [Google Scholar]
- 37.Miller FG, Joffe S: Benefit in phase 1 oncology trials: Therapeutic misconception or reasonable treatment option?. Clin Trials 5:617-623, 2008 [DOI] [PubMed] [Google Scholar]
- 38.Schupmann W, Li X, Wendler D: Do the potential medical benefits of phase 1 pediatric oncology trials justify the risks? Views of the United States public. J Pediatr 238:249-258.e3, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Palmer AC, Sorger PK: Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell 171:1678-1691.e13, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Carlisle BG, Doussau A, Kimmelman J: Benefit, burden, and impact for a cohort of post-approval cancer combination trials. Clin Trials 17:18-29, 2020 [DOI] [PubMed] [Google Scholar]
- 41.Minard-Colin V, Auperin A, Pillon M, et al. : Rituximab for high-risk, mature B-cell non-Hodgkin's lymphoma in children. N Engl J Med 382:2207-2219, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Castellino SM, Pei Q, Parsons SK, et al. : Brentuximab vedotin demonstrates superior event-free survival (EFS) in children with newly diagnosed high-risk Hodgkin lymphoma (HL): A report from the children's oncology group phase 3 study AHOD1331 (NCT 02166463). ASCO Annu Meet, 2022. Abstr #7504
- 43.Kutny MA, Alonzo TA, Abla O, et al. : Assessment of arsenic trioxide and all-trans retinoic acid for the treatment of pediatric acute promyelocytic leukemia: A report from the children's oncology group AAML1331 trial. JAMA Oncol 8:79-87, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Brown PA, Ji L, Xu X, et al. : Effect of Postreinduction therapy consolidation with blinatumomab vs chemotherapy on disease-free survival in children, adolescents, and young adults with first relapse of B-cell acute lymphoblastic leukemia: A randomized clinical trial. JAMA 325:833-842, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Locatelli F, Zugmaier G, Rizzari C, et al. : Effect of blinatumomab vs chemotherapy on event-free survival among children with high-risk first-relapse B-cell acute lymphoblastic leukemia: A randomized clinical trial. JAMA 325:843-854, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dunsmore KP, Winter SS, Devidas M, et al. : Children's oncology group AALL0434: A phase III randomized clinical trial testing nelarabine in newly diagnosed T-cell acute lymphoblastic leukemia. J Clin Oncol 38:3282-3293, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Schultz KR, Devidas M, Bowman WP, et al. : Philadelphia chromosome-negative very high-risk acute lymphoblastic leukemia in children and adolescents: Results from children's oncology group study AALL0031. Leukemia 28:964-967, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Shen S, Chen X, Cai J, et al. : Effect of dasatinib vs imatinib in the treatment of pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: A randomized clinical trial. Clinical trial. JAMA Oncol 6:358-366, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Mody R, Naranjo A, Van Ryn C, et al. : Irinotecan-temozolomide with temsirolimus or dinutuximab in children with refractory or relapsed neuroblastoma (COG ANBL1221): An open-label, randomised, phase 2 trial. Lancet Oncol 18:946-957, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dolan ME, Mitchell RB, Mummert C, et al. : Effect of O6-benzylguanine analogues on sensitivity of human tumor cells to the cytotoxic effects of alkylating agents. Cancer Res 51:3367-3372, 1991 [PubMed] [Google Scholar]
- 51.Friedman HS, Pluda J, Quinn JA, et al. : Phase I trial of carmustine plus O6-benzylguanine for patients with recurrent or progressive malignant glioma. J Clin Oncol 18:3522-3528, 2000 [DOI] [PubMed] [Google Scholar]
- 52.Blumenthal DT, Rankin C, Stelzer KJ, et al. : A Phase III study of radiation therapy (RT) and O6-benzylguanine + BCNU versus RT and BCNU alone and methylation status in newly diagnosed glioblastoma and gliosarcoma: Southwest Oncology Group (SWOG) study S0001. Int J Clin Oncol 20:650-658, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Italiano A, Infante JR, Shapiro GI, et al. : Phase I study of the checkpoint kinase 1 inhibitor GDC-0575 in combination with gemcitabine in patients with refractory solid tumors. Ann Oncol 29:1304-1311, 2018 [DOI] [PubMed] [Google Scholar]
- 54.Sun C, Hobor S, Bertotti A, et al. : Intrinsic resistance to MEK inhibition in KRAS mutant lung and colon cancer through transcriptional induction of ERBB3. Cel Rep 7:86-93, 2014 [DOI] [PubMed] [Google Scholar]
- 55.Huijberts SCFA, van Geel RMJM, van Brummelen EMJ, et al. : Phase I study of lapatinib plus trametinib in patients with KRAS-mutant colorectal, non-small cell lung, and pancreatic cancer. Cancer Chemother Pharmacol 85:917-930, 2020 [DOI] [PubMed] [Google Scholar]
- 56.Brummelen EM, Huijberts S, Herpen C, et al. : Phase I study of afatinib and selumetinib in patients with KRAS-mutated colorectal, non-small cell lung, and pancreatic cancer. Oncologist 26:290-e545, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.van Geel RMJM, van Brummelen EMJ, Eskens FALM, et al. : Phase 1 study of the pan-HER inhibitor dacomitinib plus the MEK1/2 inhibitor PD-0325901 in patients with KRAS-mutation-positive colorectal, non-small-cell lung and pancreatic cancer. Br J Cancer 122:1166-1174, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Biondi A, Gandemer V, De Lorenzo P, et al. : Imatinib treatment of paediatric Philadelphia chromosome-positive acute lymphoblastic leukaemia (EsPhALL2010): A prospective, intergroup, open-label, single-arm clinical trial. Lancet Haematol 5:e641-e652, 2018 [DOI] [PubMed] [Google Scholar]
- 59.Knörr F, Schellekens KPJ, Schoot RA, et al. : Combination Therapy with Crizotinib and Vinblastine for Relapsed or Refractory Pediatric ALK-Positive Anaplastic Large Cell Lymphoma. Haematologica 108:1442- 1446, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.O'Neil NJ, Bailey ML, Hieter P: Synthetic lethality and cancer. Nat Rev Genet 18:613-623, 2017 [DOI] [PubMed] [Google Scholar]
- 61.Innovative Therapies for Children with Cancer—Pediatric Preclinical Proof-Of Concept Project. https://www.itccp4.eu [Google Scholar]
- 62.NCI-Funded Pediatric Preclinical Testing Consortium (PPTC). http://www.ncipptc.org [Google Scholar]
- 63.Geoerger B, Bouffet E, Whitlock JA, et al. : Dabrafenib + trametinib combination therapy in pediatric patients with BRAF V600-mutant low-grade glioma: Safety and efficacy results. J Clin Oncol 38:10506, 2020. (supp; abstr 10506)NCT02124772 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Fouladi M, Perentesis JP, Wagner LM, et al. : A phase I study of cixutumumab (IMC-A12) in combination with temsirolimus (CCI-779) in children with recurrent solid tumors: A children's oncology group phase I consortium report. Clin Cancer Res 21:1558-1565, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Schafer ES, Rau RE, Berg SL, et al. : Phase 1/2 trial of talazoparib in combination with temozolomide in children and adolescents with refractory/recurrent solid tumors including ewing sarcoma: A children's oncology group phase 1 consortium study (ADVL1411). Pediatr Blood Cancer 67:e28073, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Dose Escalation and Cohort Expansion Study of Niraparib and Dostarlimab in Pediatric Participants with Solid Tumors (SCOOP). https://clinicaltrials.gov/ct2/show/NCT04544995 [Google Scholar]
- 67.Study of the Bromodomain (BRD) and Extra-terminal Domain (BET) Inhibitor BMS-986158 in Pediatric Cancer. https://clinicaltrials.gov/ct2/show/NCT03936465 [Google Scholar]
- 68.Desai AV, Robinson GW, Gauvain K, et al. : Entrectinib in children and young adults with solid or primary CNS tumors harboring NTRK, ROS1 or ALK aberrations (STARTRK-NG). Neuro-Oncology 24:1776-1789, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.O'Quigley J, Pepe M, Fisher L: Continual reassessment method: A practical design for phase 1 clinical trials in cancer. Biometrics 46:33-48, 1990 [PubMed] [Google Scholar]
- 70.Casanova M, Bautista F, Campbell Hewson Q, et al. : Phase I study of regorafenib in combination with vincristine and irinotecan in pediatric patients with recurrent or refractory solid tumors. J Clin Oncol 38:10507, 2020. (supp; abstr 10507)NCT02085148 [Google Scholar]
- 71.Iasonos A, O'Quigley J: Randomised Phase 1 clinical trials in oncology. Br J Cancer 125:920-926, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Stathis A, Iasonos A, Seymour JF, et al. : Report of the 14th international conference on malignant lymphoma (ICML) closed Workshop on future design of clinical trials in lymphomas. Clin Cancer Res 24:2993-2998, 2018 [DOI] [PubMed] [Google Scholar]
- 73.Olmos D, Postel-Vinay S, Molife LR, et al. : Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R antibody figitumumab (CP-751, 871) in patients with sarcoma and ewing’s sarcoma: A phase 1 expansion cohort study. Lancet Oncol 11:129-135, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Foster JH, Voss SD, Hall DC, et al. : Activity of crizotinib in patients with ALK-aberrant relapsed/refractory neuroblastoma: A children's oncology group study (ADVL0912). Clin Cancer Res 27:3543-3548, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ped-PRO-CTCAE/Ped-PRO-CTCAE [Caregiver] : Instrument & Form Builder. https://healthcaredelivery.cancer.gov/pro-ctcae/instrument-ped.html [Google Scholar]
- 76.Hinds PS, Pinheiro LC, McFatrich M, et al. : Recommended scoring approach for the pediatric patient-reported outcomes version of the Common Terminology Criteria for Adverse Events. Pediatr Blood Cancer 69:e29452, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Freyer DR, Lin L, Mack JW, et al. : Lack of concordance in symptomatic adverse event reporting by children, clinicians, and caregivers: Implications for cancer clinical trials. J Clin Oncol 40:1623-1634, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wages NA, Conaway MR, O'Quigley J: Dose-finding design for multi-drug combinations. Clin Trials 8:380-389, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Skolnik JM, Barrett JS, Jayaraman B, et al. : Shortening the timeline of pediatric phase I trials: The rolling six design. J Clin Oncol 26:190-195, 2008 [DOI] [PubMed] [Google Scholar]
- 80.Saha PT, Fine JP, Ivanova A: Consistency of the CRM when the dose-toxicity curve is not monotone and its application to the POCRM. Stat Med 40:2073-2082, 2021 [DOI] [PubMed] [Google Scholar]
- 81.Fraisse J, Dinart D, Tosi D, et al. : Optimal biological dose: A systematic review in cancer phase I clinical trials. BMC Cancer 21:60, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bautista F, Moreno L, Marshall L, et al. : Revisiting the definition of dose-limiting toxicities in paediatric oncology phase I clinical trials: An analysis from the Innovative Therapies for Children with Cancer Consortium. Eur J Cancer 86:275-284, 2017 [DOI] [PubMed] [Google Scholar]
- 83.Ratain MJ, Goldstein DA, Lichter AS: Interventional pharmacoeconomics—A new discipline for a cost-constrained environment. JAMA Oncol 5:1097-1098, 2019 [DOI] [PubMed] [Google Scholar]
- 84.Serritella AV, Strohbehn GW, Goldstein DA, et al. : Interventional pharmacoeconomics: A novel mechanism for Unlocking value. Clin Pharmacol Ther 108:487-493, 2020 [DOI] [PubMed] [Google Scholar]
- 85.Project Optimus—Reforming the dose optimization and dose selection paradigm in oncology. www.fda.gov/about-fda/oncology-center-excellence/project-optimus [Google Scholar]
- 86.Shah M, Rahman A, Theoret MR, et al. : The drug-dosing conundrum in oncology—When less is more. N Engl J Med 385:1445-1447, 2021 [DOI] [PubMed] [Google Scholar]
- 87.Goldsmith KC, Verschuur A, Morgenstern DA, et al. : The first report of pediatric patients with solid tumors treated with venetoclax. J Clin Oncol 38:10524, 2020. (supp; abstr 10524) [Google Scholar]
- 88.Place AE, Goldsmith K, Bourquin J-P, et al. : Accelerating drug development in pediatric cancer: A novel phase I study design of venetoclax in relapsed/refractory malignancies. Future Oncol 14:2115-2129, 2018 [DOI] [PubMed] [Google Scholar]
- 89.Codevelopment of Two or More New Investigational Drugs for Use in Combination. https://www.fda.gov/files/drugs/published/Codevelopment-of-Two-or-More-New-Investigational-Drugs-for-Use-in-Combination.pdf [Google Scholar]
- 90.https://www.ema.europa.eu/en/documents/pip-decision/p/0217/2021-ema-decision-9-june-2021-acceptance-modification-agreed-paediatric-investigation-plan_en.pdf European Medicines Agency: European Medicines Agency decision P/0217/2021 of 9 June 2021 on the acceptance of a modification of an agreed paediatric investigation plan for venetoclax (Venclyxto), (EMEA-002018-PIP02-16-M04) in accordance with Regulation (EC) No 1901/2006 of the European Parliament and of the Council.
- 91.A Study of LY3295668 Erbumine in Participants with Relapsed/Refractory Neuroblastoma. https://clinicaltrials.gov/ct2/show/NCT04106219 [Google Scholar]
- 92.Rubinstein LV, Korn EL, Freidlin B, et al. : Design issues of randomized phase II trials and a proposal for phase II screening trials. J Clin Oncol 23:7199-7206, 2005 [DOI] [PubMed] [Google Scholar]
- 93.Torres-Saavedra PA, Winter KA: An Overview of phase 2 clinical trial designs. Int J Radiat Oncol*Biol*Physics 112:22-29, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Billingham L, Malottki K, Steven N: Research methods to change clinical practice for patients with rare cancers. Lancet Oncol 17:e70-e80, 2016 [DOI] [PubMed] [Google Scholar]
- 95.Jung SH: Randomized phase II trials with a prospective control. Stat Med 27:568-583, 2008 [DOI] [PubMed] [Google Scholar]
- 96.Mandrekar SJ, Sargent DJ: Pick the winner designs in phase II cancer clinical trials. J Thorac Oncol 1:5-6, 2006 [PubMed] [Google Scholar]
- 97.Korn EL, Freidlin B: Outcome—Adaptive randomization: Is it useful?. J Clin Oncol 29:771-776, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Del Paggio JC, Berry JS, Hopman WM, et al. : Evolution of the randomized clinical trial in the era of precision oncology. JAMA Oncol 7:728-734, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ensign LG, Gehan EA, Kamen DS, et al. : An optimal three-stage design for phase II clinical trials. Stat Med 13:1727-1736, 1994 [DOI] [PubMed] [Google Scholar]
- 100.Simon R, Christian MC, Rubinstein L, et al. : Accelerated titration designs for phase I clinical trials in oncology. JNCI J Natl Cancer Inst 89:1138-1147, 1997 [DOI] [PubMed] [Google Scholar]
- 101.Gaspar N, Venkatramani R, Hecker-Nolting S, et al. : Lenvatinib with etoposide plus ifosfamide in patients with refractory or relapsed osteosarcoma (ITCC-050): A multicentre, open-label, multicohort, phase 1/2 study. Lancet Oncol 22:1312-1321, 2021 [DOI] [PubMed] [Google Scholar]
- 102.Gaspar N, Campbell-Hewson Q, Gallego Melcon S, et al. : Phase I/II study of single-agent lenvatinib in children and adolescents with refractory or relapsed solid malignancies and young adults with osteosarcoma (ITCC-050). ESMO Open 6:100250, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Vo KT, Michlitsch JG, Shah AT, et al. : Phase I trial of pazopanib in combination with irinotecan and temozolomide (PAZIT) for children and young adults with advanced sarcoma. J Clin Oncol 38:10526, 2020. (supp 6; abstr 10526)NCT03139331 [Google Scholar]
- 104.DuBois SG, Marachelian A, Fox E, et al. : Phase I study of the aurora A kinase inhibitor alisertib in combination with irinotecan and temozolomide for patients with relapsed or refractory neuroblastoma: A NANT (new approaches to neuroblastoma therapy) trial. J Clin Oncol 34:1368-1375, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mody R, Naranjo A, Van Ryn C, et al. : Irinotecan-temozolomide with temsirolimus or dinutuximab in children with refractory or relapsed neuroblastoma (COG ANBL1221): An open-label, randomised, phase 2 trial. Lancet Oncol 18:946-957, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.DuBois SG, Groshen S, Park JR, et al. : Phase I study of vorinostat as a radiation sensitizer with 131I-metaiodobenzylguanidine (131I-MIBG) for patients with relapsed or refractory neuroblastoma. Clin Cancer Res 21:2715-2721, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Fouladi M, Park JR, Stewart CF, et al. : Pediatric phase I trial and pharmacokinetic study of vorinostat: A children's oncology group phase I consortium report. J Clin Oncol 28:3623-3629, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Durvalumab and Tremelimumab for Pediatric Malignancies. https://clinicaltrials.gov/ct2/show/NCT03837899 [Google Scholar]
- 109.Dela Cruz FS, DuBois SG, Fox E, et al. : A phase I/II study of lenvatinib (LEN) plus everolimus (EVE) in recurrent and refractory pediatric solid tumors, including CNS tumors. J Clin Oncol 38:10527, 2020. (supp 6; abstr 10527) [Google Scholar]
- 110.ClinicalTrials.gov . https://clinicaltrials.gov/
- 111.FaR-RMS : An Overarching Study for Children and Adults with Frontline and Relapsed RhabdoMyoSarcoma (FaR-RMS). https://clinicaltrials.gov/ct2/show/NCT04625907 [Google Scholar]
- 112.A Study to Compare the Efficacy and Safety of Ifosfamide and Etoposide with or without Lenvatinib in Children, Adolescents and Young Adults with Relapsed and Refractory Osteosarcoma. https://clinicaltrials.gov/ct2/show/NCT04154189 [Google Scholar]
- 113.Bouffet E, Hansford J, Garre ML, et al. : Primary analysis of a phase II trial of dabrafenib plus trametinib (dab + tram) in BRAF V600–mutant pediatric low-grade glioma (pLGG). J Clin Oncol:40, 2022. 34388022 [Google Scholar]





