18.12.2012 Views

2012 EDUCATIONAL BOOK - American Society of Clinical Oncology

2012 EDUCATIONAL BOOK - American Society of Clinical Oncology

2012 EDUCATIONAL BOOK - American Society of Clinical Oncology

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

COMBINING TARGETED AGENTS IN CLINICAL TRIALS<br />

the doses for combination can occur in many permutations.<br />

Dose-escalation rules for combination therapy must take into<br />

consideration preclinical data for the single agent and the<br />

combination. Depending on the stage <strong>of</strong> development <strong>of</strong> each<br />

agent, their mechanism <strong>of</strong> action and the potential overlapping<br />

toxicities, different scenarios are envisioned: dose escalation<br />

is probably best pursued sequentially, increasing the<br />

dose <strong>of</strong> one compound in every step; dose escalation <strong>of</strong> both<br />

drugs at the same time is not recommended; and to optimize<br />

antitumor cell kill there is also merit in fixing the drug<br />

targeting the driver pathway (e.g., BRAF/MEK inhibitor for<br />

BRAF mutation in melanoma) at the highest tolerable dose<br />

while dose escalating the combination drug (e.g., PI3K/AKT/<br />

TOR inhibitor). When combining two drugs with no pharmacokinetic<br />

interactions and nonoverlapping mechanisms<br />

<strong>of</strong> action and toxicity, and when we already have extensive<br />

experience regarding the tolerability pr<strong>of</strong>ile <strong>of</strong> each drug,<br />

fewer dose-escalation steps may be necessary to accelerate<br />

combination development, as recently reported in a trial<br />

exploring the combination <strong>of</strong> sunitinib and everolimus in<br />

metastatic renal cell carcinoma (mRCC). 24<br />

Novel combinatory designs have also been suggested as<br />

potential improvements over algorithmic approaches. These<br />

strategies try to pursue fewer levels <strong>of</strong> dose escalation,<br />

thereby reducing the rate <strong>of</strong> patients exposed to biologically<br />

inactive doses without exposing them to more toxicities, in<br />

contrast to more classical drug development designs. 25<br />

Novel models <strong>of</strong> multiple parallel cohorts permit escalating<br />

two or more drugs in a sequential/parallel manner based on<br />

mechanism-related toxicities. 26 Bayesian model-based designs<br />

are based on continual reassessment <strong>of</strong> risk <strong>of</strong> toxicities<br />

and have been applied to dose escalation <strong>of</strong> combination<br />

targeted agents. 27<br />

Flexible dose escalation and de-escalation rules and intense<br />

knowledge about the mechanisms <strong>of</strong> toxicity <strong>of</strong> each<br />

drug in the combination are required to maximize outcomes<br />

without compromising a potentially or already known active<br />

dose <strong>of</strong> a single agent. Despite the availability <strong>of</strong> these novel<br />

trial designs, they are not commonly used.<br />

Furthermore, although the rationale for the combination<br />

should consider which pathway is the main driver <strong>of</strong> the<br />

disease and therefore which drug is administered at a dose<br />

level closer to its single-agent MTD, investigators may have<br />

to be prepared to decrease the dose <strong>of</strong> the main drug if the<br />

biologic rationale <strong>of</strong> a trial suggests a likelihood that synergism<br />

and full-dose combination is not tolerable.<br />

Biomarker-Guided Selection <strong>of</strong> Patients<br />

Although the selection <strong>of</strong> patients in early-phase trials<br />

based on biomarkers may make trial recruitment more difficult,<br />

combinations <strong>of</strong> targeted agents based on hypotheses <strong>of</strong><br />

synergistic biologic effect will ultimately require patient selection<br />

using biomarkers that identify populations that may<br />

benefit. Unfortunately, <strong>of</strong>ten the development <strong>of</strong> valid biomarkers<br />

that predict for benefit are <strong>of</strong>ten slow to develop<br />

in both clinical and preclinical models, making patient selection<br />

in early-phase trials difficult. The development <strong>of</strong> predictive<br />

biomarkers for patient selection is crucial to the success <strong>of</strong><br />

targeted agents as well as combinations <strong>of</strong> targeted agents.<br />

Assessment <strong>of</strong> Tolerability in Combination Studies<br />

Since chronic dosing is usually indicated for these drug<br />

combinations, delayed and cumulative toxicities should be<br />

carefully considered when selecting the recommended dose<br />

for further development. As an example, a phase I trial <strong>of</strong><br />

the combination <strong>of</strong> bevacizumab and everolimus 28 escalated<br />

doses up to 10 mg/kg fortnightly and 10 mg/d po respectively,<br />

without observing dose-limiting toxicities during the<br />

first cycles <strong>of</strong> treatment. In a phase II trial in mRCC, 29 up to<br />

a quarter <strong>of</strong> the patients had grade 3 or 4 proteinuria with<br />

the combination, while grade 3 or 4 proteinuria with bevacizumab<br />

alone at the same dose in mRCC was below 8%. 30<br />

Trial designs that integrate information from late onset<br />

toxicities for deciding regarding dose selection may be warranted.<br />

31 In addition, clinical trials should aim to assess<br />

mechanistic data to define causal relationships with drug<br />

and observed toxicities to make better informed decisions<br />

regarding choices <strong>of</strong> combination targeted agents. Uncertainty<br />

regarding the mechanisms behind targeted agent<br />

toxicity and <strong>of</strong>f-target drug effects may negatively affect<br />

promising combinations.<br />

Regulatory Concerns and Legal Barriers<br />

The evaluation <strong>of</strong> different combinations <strong>of</strong> several targeted<br />

agents from multiple companies may sometimes be<br />

challenging because <strong>of</strong> competing interests, and may be<br />

further complicated if multiple sponsors are not fully committed<br />

to the trial. A clear example may be combining an<br />

established drug or chemotherapy regimen with a novel<br />

agent, as the outcome <strong>of</strong> the trial may not be equally<br />

pr<strong>of</strong>itable for both sponsors. Apart from intellectual property<br />

issues and the requirement <strong>of</strong> financial support, the<br />

risk <strong>of</strong> novel toxicities and adverse outcomes with concomitant<br />

administration might be considered a risky commercial<br />

decision for sponsors whose antitumor compound is already<br />

at a more advanced stage <strong>of</strong> development. Academic institutions,<br />

patient lobbies, and regulatory authorities may<br />

need to assume a championing role for some combinations<br />

with a strong biologic rationale that are perhaps less commercially<br />

attractive.<br />

Conclusion<br />

The advent <strong>of</strong> targeted therapies has shifted the emphasis<br />

in drug development from cytotoxic chemotherapy combinations<br />

to selective and potent molecular therapeutics. Such<br />

specific inhibitors may not result in significant or durable<br />

antitumor activity as single agents, and should therefore be<br />

given in combination with one or more drugs to molecularly<br />

defined populations <strong>of</strong> patients. We should now be focusing<br />

more efforts on combinatorial drug development strategies,<br />

perhaps even from the outset <strong>of</strong> first-in-human trials to<br />

maximize the likelihood <strong>of</strong> benefit to patients dying <strong>of</strong><br />

advanced cancer. Such an approach may be able to further<br />

accelerate the delivery <strong>of</strong> anticancer treatment to benefit<br />

and improve the lives <strong>of</strong> our patients with cancer.<br />

ACKNOWLEDGMENTS<br />

The Drug Development Unit <strong>of</strong> the Royal Marsden NHS<br />

Foundation Trust and The Institute <strong>of</strong> Cancer Research is<br />

supported in part by a program grant from Cancer Research<br />

United Kingdom. Support was also provided by the Experimental<br />

Cancer Medicine Centre (to The Institute <strong>of</strong> Cancer<br />

Research) and the National Institute for Health Research Biomedical<br />

Research Centre (jointly to the Royal Marsden NHS<br />

Foundation Trust and The Institute <strong>of</strong> Cancer Research).<br />

673

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!