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 rational combination <strong>of</strong> mTOR inhibitors with either AKT<br />

or PIK3CA inhibitors or the development <strong>of</strong> TOR kinase<br />

inhibitors. This strategy may be best utilized in tumors driven<br />

mainly by one pathway but which may fail if alternative<br />

pathways are prominent as a mechanism <strong>of</strong> resistance. This<br />

strategy also has the disadvantage <strong>of</strong> possible overlapping<br />

drug-related toxicities via inhibiting the same pathway.<br />

Targeting parallel or unrelated pathways. This strategy<br />

maximizes the likelihood <strong>of</strong> avoiding resistance mechanisms,<br />

especially those caused by the upregulation <strong>of</strong> parallel<br />

pathways, and also minimizes overlapping toxicities. The<br />

concomitant inhibition <strong>of</strong> two pathways may also result in<br />

selective tumor cell cytotoxicity, whereas each agent alone<br />

may not be active as a single agent.<br />

An example <strong>of</strong> this approach is the dual targeting <strong>of</strong> the<br />

PI3K and Ras/Raf/Mek/ERK pathways. Deregulation <strong>of</strong> the<br />

Ras/Raf/Mek/ERK pathway has been identified as a determinant<br />

<strong>of</strong> resistance to PI3K-Akt inhibitors. 10 There is also<br />

strong preclinical evidence that ERK-independent mechanisms<br />

<strong>of</strong> resistance to B-RAF and MEK inhibitors are<br />

mediated by the upregulation <strong>of</strong> the PIK3CA/Akt/mTOR<br />

pathway. 11 In murine models, double inhibition <strong>of</strong> PIK3CA<br />

and MEK shows synergistic antitumor activity for K-RAS<br />

mutant tumors, 12 and PI3K-Akt activation leads to MEKinhibitor<br />

resistance in K-RAS mutants, which is reversible<br />

when blocking both pathways. 13 Moreover, mutations in<br />

both pathways tend to coexist. 14<br />

<strong>Clinical</strong> trials are currently being conducted to evaluate<br />

this strategy <strong>of</strong> targeting parallel pathways. Recently, a<br />

first-in-human trial <strong>of</strong> the pan-AKT inhibitor MK-2206 has<br />

been reported; although strong pharmacodynamics evidence<br />

<strong>of</strong> AKT signaling blockade was demonstrated, minimal evidence<br />

<strong>of</strong> single-agent antitumor activity was observed. 15 A<br />

combination trial <strong>of</strong> MK-2206 with the MEK inhibitor<br />

AZD6244 is currently being evaluated (NCT01021748) and<br />

is demonstrating antitumor activity in KRAS-mutant cancers.<br />

Other similar early-phase trials <strong>of</strong> combinations based<br />

on this approach include drugs targeting MEK and mTOR<br />

(NCT01378377), or MEK and PIK3CA/mTOR inhibition<br />

KEY POINTS<br />

● The biology <strong>of</strong> cancer cells involves complex signaling<br />

networks, so anticancer strategies may not be successful<br />

if blocking single targets.<br />

● A combination <strong>of</strong> targeted agents will be necessary to<br />

maximize antitumor activity, counteracting primary<br />

and secondary resistance.<br />

● The rationale for evaluating combinations <strong>of</strong> drugs<br />

must rely on biology-driven hypothesis and smart<br />

screening strategies.<br />

● Different strategies include using two or more drugs<br />

against the same target, targeting different points<br />

upstream and downstream <strong>of</strong> one pathway, or targeting<br />

several pathways simultaneously on which the<br />

cancer cell is dependent.<br />

● Design <strong>of</strong> clinical trials with combinations <strong>of</strong> targeted<br />

agents should consider how to deal with the challenges<br />

pertaining to such studies.<br />

Table 1. Opportunities and Pitfalls <strong>of</strong> Combining<br />

Targeted Agents<br />

Opportunities Pitfalls<br />

1. Validate novel biologic hypotheses<br />

2. Synergize antitumor effect without<br />

synergizing toxicity: increasing the<br />

therapeutic window<br />

3. Further develop single agents that do<br />

not have activity as monotherapy:<br />

synthetic lethality<br />

4. Counteract primary and secondary<br />

resistance<br />

5. Develop novel indications for existing<br />

and/or approved drugs<br />

(NCT01390818). These data allow us to envision the testing<br />

<strong>of</strong> biology-driven hypotheses with multiple combinations;<br />

these may eventually require more drugs for optimal blockade<br />

<strong>of</strong> cancer growth. Combinations for the treatment <strong>of</strong><br />

advanced prostate cancer that we propose include abiraterone<br />

acetate and MDV3100; either <strong>of</strong> these agents with<br />

PI3K/AKT/TOR inhibitors; or possibly triple therapy with<br />

abiraterone, MDV3100, and a PI3K/TOR inhibitor.<br />

Choosing the Right Combination Therapy<br />

1. Unreliable preclinical models<br />

2. Optimal selection <strong>of</strong> drugs and<br />

targets to combine<br />

3. Optimal sequence and dosage <strong>of</strong><br />

the combination<br />

4. Risk <strong>of</strong> overlapping toxicities<br />

5. Lack <strong>of</strong> standardized design for<br />

phase I/II trials for combination<br />

targeted therapies<br />

6. Competing interests <strong>of</strong> different<br />

researchers, corporations, and/or<br />

institutions to combine therapies<br />

When combining two (or more) targeted therapies for the<br />

first time, each must be tested at various doses and dosing<br />

schedules in a phase I study. However, the process <strong>of</strong> dose<br />

finding, or defining the recommended phase II trial dose,<br />

can take 1 year or longer to complete depending on the<br />

complexity <strong>of</strong> the trial and compatibility <strong>of</strong> the agents, all at<br />

significant cost. Therefore, it is critical to utilize strategies<br />

that identify the best therapeutic combinations, and furthermore<br />

identify targets that cancer cells are reliant on so that<br />

when blocked, there is a lethal or at least cytostatic effect.<br />

This type <strong>of</strong> approach is based on concepts <strong>of</strong> oncogene<br />

addition, nononcogene addiction, and synthetic lethality<br />

that attack the “hallmarks” <strong>of</strong> cancer. 16<br />

Adopting systems-based approaches may help to address<br />

this complexity, such as utilizing information from deep<br />

transcriptomic, genomic, proteomic, and/or metabolic analyses.<br />

17 Although this “personalized” approach is attractive,<br />

it may <strong>of</strong>ten yield information on targets for which there is<br />

no active drug, for which there are only “passenger” mutations<br />

rather than drivers <strong>of</strong> oncogenesis, or for genes for<br />

which we have incomplete functional information. It has<br />

been suggested that systems-based approaches can be used<br />

to select targeted combinations 17 ; however, further development<br />

and application <strong>of</strong> bioinformatics is clearly necessary<br />

to process the high amounts <strong>of</strong> data derived from massive<br />

screening strategies.<br />

Another strategy is to identify potential drug combinations<br />

using RNA interference (RNAi) to simulate models <strong>of</strong><br />

pharmacologic inhibition. In this method, wild-type cell lines<br />

or cell lines with induced loss <strong>of</strong> function for a particular<br />

target are exposed to RNAi libraries with or without exposure<br />

to a particular drug to look for antitumoral synergism<br />

or drug sensitization. These RNAi screens can therefore<br />

select potential combination <strong>of</strong> targets to be tested further<br />

18,19 and ideally identify potential synthetic lethality<br />

effects when inhibiting two a priori nonlethal targets. How-<br />

671

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

Saved successfully!

Ooh no, something went wrong!