Showing posts with label pharma legislation. Show all posts
Showing posts with label pharma legislation. Show all posts

Wednesday, February 22, 2017

Establishing efficacy - without humans?

The decade following passage of FDAAA has been one of easing standards for drug approvals in the US, most notably with the advent of “breakthrough” designation created by FDASIA in 2012 and the 21st Century Cures Act in 2016.

Although, as of this writing, there is no nominee for FDA Commissioner, it appears to be safe to say that the current administration intends to accelerate the pace of deregulation, mostly through further lowering of approval requirements. In fact, some of the leading contenders for the position are on record as supporting a return to pre-Kefauver-Harris days, when drug efficacy was not even considered for approval.
Build a better mouse model, and pharma will
beat a path to your door - no laws needed.

In this context, it is at least refreshing to read a proposal to increase efficacy standards. This comes from two bioethicists at McGill University, who make the somewhat-startling case for a higher degree of efficacy evaluation before a drug begins any testing in humans.
We contend that a lack of emphasis on evidence for the efficacy of drug candidates is all too common in decisions about whether an experimental medicine can be tested in humans. We call for infrastructure, resources and better methods to rigorously evaluate the clinical promise of new interventions before testing them on humans for the first time.
The author propose some sort of centralized clearinghouse to evaluate efficacy more rigorously. It is unclear what they envision this new multispecialty review body’s standards for green-lighting a drug to enter human testing. Instead they propose three questions:
  • What is the likelihood that the drug will prove clinically useful?
  • Assume the drug works in humans. What is the likelihood of observing the preclinical results?
  • Assume the drug does not work in humans. What is the likelihood of observing the preclinical results?
These seem like reasonable questions, I suppose – and are likely questions that are already being asked of preclinical data. They certainly do not rise to the level of providing a clear standard for regulatory approval, though perhaps it’s a reasonable place to start.

The most obvious counterargument here is one that the authors curiously don’t pick up on at all: if we had the ability to accurately (or even semiaccurately) predict efficacy preclinically, pharma sponsors would already be doing it. The comment notes: “More-thorough assessments of clinical potential before trials begin could lower failure rates and drug-development costs.” And it’s hard not to agree: every pharmaceutical company would love to have even an incrementally-better sense of whether their early pipeline drugs will be shown to work as hoped.

The authors note
Commercial interests cannot be trusted to ensure that human trials are launched only when the case for clinical potential is robust. We believe that many FIH studies are launched on the basis of flimsy, underscrutinized evidence.
However, they do not produce any evidence that industry is in any way deliberately underperforming their preclinical work, merely that preclinical efficacy is often difficult to reproduce and is poorly correlated with drug performance in humans.

Pharmaceutical companies have many times more candidate compounds than they can possibly afford to put into clinical trials. Figuring out how to lower failure rates – or at least the total cost of failure - is a prominent industry obsession, and efficacy remains the largest source of late-stage trial failure. This quest to “fail faster” has resulted in larger and more expensive phase 2 trials, and even to increased efficacy testing in some phase 1 trials. And we do this not because of regulatory pressure, but because of hopes that these efforts will save overall costs. So it seems beyond probable that companies would immediately invest more in preclinical efficacy testing, if such testing could be shown to have any real predictive power. But generally speaking, it does not.

As a general rule, we don’t need regulations that are firmly aligned with market incentives, we need regulations if and when we think those incentives might run counter to the general good. In this case, there are already incredibly strong market incentives to improve preclinical assessments. Where companies have attempted to do something with limited success, it would seem quixotic to think that regulatory fiat will accomplish more.

(One further point. The authors try to link the need for preclinical efficacy testing to the 2016 Bial tragedy. This seems incredibly tenuous: the authors speculate that perhaps trial participants would not have been harmed and killed if Bial had been required to produce more evidence of BIA102474’s clinical efficacy before embarking on their phase 1 trials. But that would have been entirely coincidental in this case: if the drug had in fact more evidence of therapeutic promise, the tragedy still would have happened, because it had nothing at all to do with the drug’s efficacy.

This is to some extent a minor nitpick, since the argument in favor of earlier efficacy testing does not depend on a link to Bial. However, I bring it up because a) the authors dedicate the first four paragraphs of their comment to the link, and b) there appears to be a minor trend of using the death and injuries of that trial to justify an array of otherwise-unrelated initiatives. This seems like a trend we should discourage.)

[Update 2/23: I posted this last night, not realizing that only a few hours earlier, John LaMattina had published on this same article. His take is similar to mine, in that he is suspicious of the idea that pharmaceutical companies would knowingly push ineffective drugs up their pipeline.]

ResearchBlogging.org Kimmelman, J., & Federico, C. (2017). Consider drug efficacy before first-in-human trials Nature, 542 (7639), 25-27 DOI: 10.1038/542025a

Wednesday, July 31, 2013

Brazen Scofflaws? Are Pharma Companies Really Completely Ignoring FDAAA?

Results reporting requirements are pretty clear. Maybe critics should re-check their methods?

Ben Goldacre has rather famously described the clinical trial reporting requirements in the Food and Drug Administration Amendments Act of 2007 as a “fake fix” that was being thoroughly “ignored” by the pharmaceutical industry.

Pharma: breaking the law in broad daylight?
He makes this sweeping, unconditional proclamation about the industry and its regulators on the basis of  a single study in the BMJ, blithely ignoring the fact that a) the authors of the study admitted that they could not adequately determine the number of studies that were meeting FDAAA requirements and b) a subsequent FDA review that identified only 15 trials potentially out of compliance, out of a pool of thousands.


Despite the fact that the FDA, which has access to more data, says that only a tiny fraction of studies are potentially noncompliant, Goldacre's frequently repeated claims that the law is being ignored seems to have caught on in the general run of journalistic and academic discussions about FDAAA.

And now there appears to be additional support for the idea that a large percentage of studies are noncompliant with FDAAA results reporting requirements, in the form of a new study in the Journal of Clinical Oncology: "Public Availability of Results of Trials Assessing Cancer Drugs in the United States" by Thi-Anh-Hoa Nguyen, et al.. In it, the authors report even lower levels of FDAAA compliance – a mere 20% of randomized clinical trials met requirements of posting results on clinicaltrials.gov within one year.

Unsurprisingly, the JCO results were immediately picked up and circulated uncritically by the usual suspects.

I have to admit not knowing much about pure academic and cooperative group trial operations, but I do know a lot about industry-run trials – simply put, I find the data as presented in the JCO study impossible to believe. Everyone I work with in pharma trials is painfully aware of the regulatory environment they work in. FDAAA compliance is a given, a no-brainer: large internal legal and compliance teams are everywhere, ensuring that the letter of the law is followed in clinical trial conduct. If anything, pharma sponsors are twitchily over-compliant with these kinds of regulations (for example, most still adhere to 100% verification of source documentation – sending monitors to physically examine every single record of every single enrolled patient - even after the FDA explicitly told them they didn't have to).

I realize that’s anecdotal evidence, but when such behavior is so pervasive, it’s difficult to buy into data that says it’s not happening at all. The idea that all pharmaceutical companies are ignoring a highly visible law that’s been on the books for 6 years is extraordinary. Are they really so brazenly breaking the rules? And is FDA abetting them by disseminating incorrect information?

Those are extraordinary claims, and would seem to require extraordinary evidence. The BMJ study had clear limitations that make its implications entirely unclear. Is the JCO article any better?

Some Issues


In fact, there appear to be at least two major issues that may have seriously compromised the JCO findings:

1. Studies that were certified as being eligible for delayed reporting requirements, but do not have their certification date listed.

The study authors make what I believe to be a completely unwarranted assumption:

In trials for approval of new drugs or approval for a new indication, a certification [permitting delayed results reporting] should be posted within 1 year and should be publicly available.

It’s unclear to me why the authors think the certifications “should be” publicly available. In re-reading FDAAA section 801, I don’t see any reference to that being a requirement. I suppose I could have missed it, but the authors provide a citation to a page that clearly does not list any such requirement.

But their methodology assumes that all trials that have a certification will have it posted:

If no results were posted at ClinicalTrials.gov, we determined whether the responsible party submitted a certification. In this case, we recorded the date of submission of the certification to ClinicalTrials.gov.

If a sponsor gets approval from FDA to delay reporting (as is routine for all drugs that are either not approved for any indication, or being studied for a new indication – i.e., the overwhelming majority of pharma drug trials), but doesn't post that approval on the registry, the JCO authors deem that trial “noncompliant”. This is not warranted: the company may have simply chosen not to post the certification despite being entirely FDAAA compliant.

2. Studies that were previously certified for delayed reporting and subsequently reported results

It is hard to tell how the authors treated this rather-substantial category of trials. If a trial was certified for delayed results reporting, but then subsequently published results, the certification date becomes difficult to find. Indeed, it appears in the case where there were results, the authors simply looked at the time from study completion to results posting. In effect, this would re-classify almost every single one of these trials from compliant to non-compliant. Consider this example trial:


  • Phase 3 trial completes January 2010
  • Certification of delayed results obtained December 2010 (compliant)
  • FDA approval June 2013
  • Results posted July 2013 (compliant)


In looking at the JCO paper's methods section, it really appears that this trial would be classified as reporting results 3.5 years after completion, and therefore be considered noncompliant with FDAAA. In fact, this trial is entirely kosher, and would be extremely typical for many phase 2 and 3 trials in industry.

Time for Some Data Transparency


The above two concerns may, in fact, be non-issues. They certainly appear to be implied in the JCO paper, but the wording isn't terribly detailed and could easily be giving me the wrong impression.

However, if either or both of these issues are real, they may affect the vast majority of "noncompliant" trials in this study. Given the fact that most clinical trials are either looking at new drugs, or looking at new indications for new drugs, these two issues may entirely explain the gap between the JCO study and the unequivocal FDA statements that contradict it.

I hope that, given the importance of transparency in research, the authors will be willing to post their data set publicly so that others can review their assumptions and independently verify their conclusions. It would be more than a bit ironic otherwise.

[Image credit: Shamless lawlessness via Flikr user willytronics.]


ResearchBlogging.org Thi-Anh-Hoa Nguyen, Agnes Dechartres, Soraya Belgherbi, and Philippe Ravaud (2013). Public Availability of Results of Trials Assessing Cancer Drugs in the United States JOURNAL OF CLINICAL ONCOLOGY DOI: 10.1200/JCO.2012.46.9577

Wednesday, August 8, 2012

Testing Transparency with the TEST Act

A quick update on my last post regarding the enormously controversial -- but completely unmentioned -- requirement to publicly report all versions of clinical trial protocols on ClinicalTrials.gov: The New England Journal of Medicine has weighed in with an editorial strongly in support of the TEST Act. 

NEJM Editor-in-Chief Jeffrey Drazen at least mentions the supporting documents requirement, but only in part of one sentence, where he confusingly refers to the act "extending results reporting to include the deposition of consent and protocol documents approved by institutional review boards." The word "deposition" does not suggest actual publication, which the act clearly requires. 

I don't think this qualifies as an improvement in transparency about the impact the TEST Act, as written, would have. I'm not surprised when a trade publication like Center Watch recycles a press release into a news item. However, it wouldn't seem like too much to ask that NEJM editorials aspire to a moderately higher standard of critical inquiry.

Monday, August 6, 2012

Public Protocols? Burying the lede on the TEST Act

Not to be confused with the Test Act.
(via Luminarium)
4 Democratic members of Congress recently co-sponsored the TEST (Trial and Experimental Studies Transparency) Act, which is intended to expand the scope of mandatory registration of clinical trials. Coverage so far has been light, and mainly consists of uncritical recycling of the press release put out by congressman Markey’s office.

Which is unfortunate, because nowhere in that release is there a single mention of the bill’s most controversial feature: publication of clinical trial "supporting documents", including the patient’s Informed Consent Form (ICF) and, incredibly, the entire protocol (including any and all subsequent amendments to the protocol).

How Rep. Markey and colleagues managed to put out a 1,000-word press release without mentioning this detail is nothing short of remarkable. Is the intent to try to sneak this through?

Full public posting of every clinical trial protocol would represent an enormous shift in how R&D is conducted in this country (and, therefore, in the entire world). It would radically alter the dynamics of how pharmaceutical companies operate by ripping out a giant chunk of every company’s proprietary investment – essentially, confiscating and nationalizing their intellectual property. 

Maybe, ultimately, that would be a good thing.  But that’s by no means clear ... and quite likely not true. Either way, however, this is not the kind of thing you bury in legislation and hope no one notices.

[Full text of the bill is here (PDF).]

[UPDATE May 17, 2013: Apparently, the irony of not being transparent with the contents of your transparency law was just too delicious to pass up, as Markey and his co-sponsors reintroduced the bill yesterday. Once again, the updated press release makes no mention of the protocol requirement.]

Tuesday, July 31, 2012

Clouding the Debate on Clinical Trials: Pediatric Edition

I would like to propose a rule for clinical trial benchmarks. This rule may appear so blindingly obvious that I run the risk of seeming simple-minded and naïve for even bringing it up.

The rule is this: if you’re going to introduce a benchmark for clinical trial design or conduct, explain its value.

Are we not putting enough resources into pediatric research, or have we over-incentivized risky experimentation on a vulnerable population?  This is a critically important question in desperate need of more data and thoughtful analysis.
That’s it.  Just a paragraph explaining the rationale of why you’ve chosen to measure what you’re measuring.  Extra credit if you compare it to other benchmarks you could have used, or consider the limitations of your new metric.

I would feel bad for bringing this up, were it not for two recent articles in major publications that completely fail to live up to this standard. I’ll cover one today and one tomorrow.

The first is a recent article in Pediatrics, Pediatric Versus Adult Drug Trials for Conditions With High Pediatric Disease Burden, which has received a fair bit of attention in the industry -- mostly due to Reuters uncritically recycling the authors’ press release

It’s worth noting that the claim made in the release title, "Drug safety and efficacy in children is rarely addressed in drug trials for major diseases", is not at all supported by any data in the study itself. However, I suppose I can live with misleading PR.  What is frustrating is the inadequacy of the measures the authors use in the actual study, and the complete lack of discussion about them.

To benchmark where pediatric drug research should be, they use the proportion of total "burden of disease" borne by children.   Using WHO estimates, they look at the ratio of burden (measured, essentially, in years of total disability) between children and adults.  This burden is further divided into high-income countries and low/middle-income countries.

This has some surface plausibility, but presents a host of issues.  Simply looking at the relative prevalence of a condition does not really give us any insights into what we need to study about treatment.  For example: number 2 on the list for middle/low income diseases is diarrheal illness, where WHO lists the burden of disease as 90% pediatric.  There is no question that diarrheal diseases take a terrible toll on children in developing countries.  We absolutely need to focus resources on improving prevention and treatment: what we do not particularly need is more clinical trials.  As the very first bullet on the WHO fact sheet points out, diarrheal diseases are preventable and treatable.  Prevention is mostly about improving the quality of water and food supplies – this is vitally important stuff, but it has nothing to do with pharmaceutical R&D.

In the US, the NIH’s National Institute for Child Health and Human Development (NICHD) has a rigorous process for identifying and prioritizing needs for pediatric drug development, as mandated by the BPCA.  It is worth noting that only 2 of the top 5 diseases in the Pediatrics article make the cut among the 41 highest-priority areas in the NICHD’s list for 2011.

(I don’t even think the numbers as calculated by the authors are even convincing on their own terms:  3 of the 5 "high burden" diseases in wealthy countries – bipolar, depression, and schizophrenia – are extremely rare in very young children, and only make this list because of their increasing incidence in adolescence.  If our objective is to focus on how these drugs may work differently in developing children, then why wouldn’t we put greater emphasis on the youngest cohorts?)

Of course, just because a new benchmark is at odds with other benchmarks doesn’t necessarily mean that it’s wrong.  But it does mean that the benchmark requires some rigorous vetting before its used.  The authors make no attempt at explaining why we should use their metric, except to say it’s "apt". The only support provided is a pair of footnotes – one of those, ironically, is to this article from 1999 that contains a direct warning against their approach:
Our data demonstrate how policy makers could be misled by using a single measure of the burden of disease, because the ranking of diseases according to their burden varies with the different measures used.
If we’re going to make any progress in solving the problems in drug development – and I think we have a number of problems that need solving – we have got to start raising our standards for our own metrics.

Are we not putting enough resources into pediatric research, or have we over-incentivized risky experimentation on a vulnerable population? This is a critically important question in desperate need of more data and thoughtful analysis. Unfortunately, this study adds more noise than insight to the debate.

Tomorrow In a couple weeks, I’ll cover the allegations about too many trials being too small. [Update: "tomorrow" took a little longer than expected. Follow up post is here.]

[Note: the Pediatrics article also uses another metric, "Percentage of Trials that Are Pediatric", that is used as a proxy for amount of research effort being done.  For space reasons, I’m not going to go into that one, but it’s every bit as unhelpful as the pediatric burden metric.]

ResearchBlogging.org Bourgeois FT, Murthy S, Pinto C, Olson KL, Ioannidis JP, & Mandl KD (2012). Pediatric Versus Adult Drug Trials for Conditions With High Pediatric Disease Burden. Pediatrics PMID: 22826574

Friday, July 13, 2012

Friday Flailing: Medical Gasses and the Law of the Excluded Middle

Buried
Aristotle never actually said "principium tertii
exclusi"
, mostly because he didn't speak Latin.
under the mass of attention and conversations surrounding the ACA last week, the FDA Safety and Innovation Act (FDASIA) contained a number of provisions that will have lasting effects in the pharmaceutical industry.  What little notice it did get tended to focus on PDUFA reauthorization and the establishment of fees for new generic drug applications (GDUFA).

(Tangent: other parts of the act are well worth looking into: NORD is happy about new incentives for rare disease research, the SWHR is happy about expanded reporting on sex and race in clinical trials, and antibiotic drug makers are happy about extra periods of market exclusivity.  A very good summary is available on the FDA Law Blog.)

So no one’s paid any attention to the Medical Gasses Safety Act, which formally defines medical gasses and even gives them their own Advisory Committee and user fees (I guess those will be MGUFAs?)

The Act’s opening definition is a bit of an eyebrow-raiser:
(2) The term ‘medical gas’ means a drug that is--
‘(A) manufactured or stored in a liquefied, non-liquefied, or cryogenic state; and
‘(B) is administered as a gas.
I’m clearly missing something here, because as far as I can tell, everything is either liquefied or non-liquefied.   This doesn’t seem to lend a lot of clarity to the definition.  And then, what to make of the third option?  How can there be a third option?  It’s been years since my college logic class, but I still remember the Law of the Excluded Middle – everything is either P or not-P. 

I was going to send an inquiry through to Congressman Leonard Lance (R-NJ), the bill’s original author, but his website regrets to inform me that he is “unable to reply to any email from constituents outside of the district.”

So I will remain trapped in Logical Limbo.  Enjoy your weekend.

Thursday, January 12, 2012

Changing the Rules, Ever So Slightly, For Rare Diseases

At the end of last year, US Reps Cliff Stearns (R-FL) and Ed Towns (D-NY) introduced the Unlocking Lifesaving Treatments for Rare-Diseases Act (ULTRA for short). Despite what its bold name might imply (and unlike many recent congressional healthcare bills), ULTRA is actually a modest and carefully-though-out piece of legislation.

The main thrust of ULTRA is to enable developers of drugs for rare diseases to take advantage of the FDA’s existing Accelerated Approval pathway. Accelerated Approval reduces the initial burden of proof for manufacturers to bring a drug to market by conducting smaller clinical trials that measure a drug’s efficacy Rare Diseases Day: Feb 29, 2012against “surrogate” endpoints – that is, endpoints that do not directly measure the disease, but rather other factors that are associated with the disease. This can greatly reduce the time and cost of clinical trials.

To qualify for Accelerated Approval, however, trials for a new drug needs to meet two conditions:

  • The drug must be studied for treatment of a serious disease, with unmet medical need

  • There must be clinical evidence that improving the surrogate endpoint is reasonably likely to predict real benefit for those with the disease

ULTRA does not change the first criterion, only the second. For rare diseases, there is often not robust clinical evidence to support surrogate endpoints, so the bill alters the language slightly to permit the FDA to accept “reasonable scientific data that support and qualify the relevance of the surrogate endpoint”. In essence, the burden to prove the validity of the surrogate has been relaxed, permitting their use in pivotal trials, and using a surrogate may reduce the number of patients needed for a trial by as much as 50-75%.

Accelerated Approval still requires the drug manufacturer to complete full trials to more firmly establish the drug’s efficacy – it just allows the drug to be available on the market while those full trials are being conducted. ULTRA does not change this requirement for drugs of rare diseases, so in the end it is not lowering the standard for these drugs at all.

Obviously, anything can happen to a bill as it wends its way through congress. But as it is currently written, ULTRA is a highly rational, well-targeted adjustment to current law that should quickly show benefits for patients with rare diseases, and deserves quick action and passage.

(Further reading: the FDA Law Blog has an excellent review of the proposed act.)