Tag Archives: progress

Tipping the balance

Almost four in ten serious adverse drug reactions now listed in the labels of 12 targeted cancer therapies were not mentioned in the studies that led to their approval. Half the serious reactions that were missed are potentially fatal. How can we improve the way we investigate and report the side effects of new drugs?

Questions are being raised about the accuracy and integrity of reports from pivotal clinical trials that provide the evidence for licensing cancer drugs. There is increasing concern that reports overstate the effectiveness of innovative drugs in a real world setting, because patients on trials are healthier and fitter than most of the people it will be used in, and understate side effects. This distorts the information used by clinicians to define the recommended dose, by regulators to assess the risk–benefit profile, and by patients to choose between treatment options.

Researchers and patient groups are calling for changes in the way that trials are designed and reported, with fewer exclusions and a much more rigorous approach to reporting side effects.

A team at the Princess Margaret Hospital in Toronto has turned a spotlight on this issue in a series of papers which highlights the gap between adverse events reported from ‘pivotal’ trials (which form the basis for marketing approval) and the warnings eventually added to drug labels – often years later.

The first of these, published in 2011, showed that 39% of serious adverse drug reactions – half of them potentially fatal – were not described in any of the randomised clinical trial (RCT) reports associated with 12 targeted anti-cancer agents (JCO 2011, 29:174–185) They had to be added to drug labels at a later date.

The same team analysed anti-cancer drugs approved by the US Food and Drug Administration (FDA) between 2000 and 2010 and found that most were associated with increased odds of toxic death, treatment discontinuation or severe adverse events (JCO 2012, 30: 3012–19).

In 2014 the team demonstrated that adverse effects also led to increased costs of treatment (JCO 2014, 32: 3634–43). Saroj Niraula, lead author on the 2012 and 2014 papers and now a medical oncologist at Cancer Care Manitoba, in Winnipeg, Canada, stresses that new therapies have saved tens of thousands of lives and that criticism of trial reports should be seen in that context. “My point is to do what we can to improve the reporting of the trials so we can make the best judgement about efficacy and toxicity, rather than pointing out flaws in reporting research.”

However, he says that RCTs are focused on demonstrating clinical efficacy rather than testing toxicity. “Frequently when we read pivotal RCT published papers we see statements like ‘no differences in toxicities’, but most trials are not powered to support such statements.”

His 2012 paper noted that treatment-related mortality associated with bevacizumab (Avastin), the cardiovascular effects of aromatase inhibitors, and the increased risk of cardiopulmonary arrest with cetuximab (Erbitux) all went unreported in the original trials.

Bevacizumab was approved in the EU for treating metastatic breast cancer in 2007 and in the US in 2008, on the basis of trial reports that showed tumour shrinkage and an increase in progression-free survival. Further evidence on both safety and efficacy that emerged in the two years following the trial, however, prompted the FDA to withdraw that approval, on the grounds that patients would “risk potentially life-threatening side effects without proof that the use of Avastin will provide a benefit, in terms of delay in tumour growth, that would justify those risks.” (The European regulators, the EMA made a different judgement call after deciding that the benefits of tumour shrinkage did outweigh the risks.)

The aromatase inhibitor Arimidex was approved by the FDA in 2002 as an adjuvant treatment for early breast cancer in postmenopausal women, on the basis of the ATAC trial, which showed improved disease-free survival compared to tamoxifen and a lower incidence of certain side effects associated with tamoxifen.

A secondary analysis of the ATAC data by the FDA later led to a warning being added to the drug label to indicate that “anastrozole may be linked to an increased risk for ischemic cardiovascular events in women with pre-existing ischemic heart disease”.
Yet the report of a ten-year update on the trial, published in 2011, made no reference to the new evidence, or the additional warning.

The 2006 trial comparing cetuximab and radiotherapy with radiotherapy alone for people with squamous carcinoma of the head and neck reported ‘similar’ incidence rates of severe reactions for the two treatment arms. The 2% of patients who died on the cetuximab arm as a result of cardiopulmonary arrest went unreported because the trial only reported acute adverse events that affected at least 10% of patients.

Lapatinib (Tykerb), is another striking example, which was flagged up by Bostjan Seruga, one of the collaborators in the ‘Toronto papers’, at a presentation he made at the European Cancer Congress in Vienna in September 2015. He pointed out that Tykerb’s drug label has been revised 12 times since it was approved in 2007 to treat women with metastatic HER2-positive breast cancer. Added warnings include notice of potential damage to lungs, severe skin reactions, and a ‘boxed’ warning on hepatotoxicity – the strongest warning that the FDA can mandate.

Seruga, who is based at the Institute of Oncology in Ljubljana, Slovenia, pointed out that new evidence from post-marketing surveillance can significantly change the risk–benefit balance, but it is the early impression about lack of harm that sticks. “Patients do not know what symptoms to expect based on prior experience, drug developers may have a false impression as to how a drug is tolerated, regulators may not have confidence in the fidelity of information about balancing risks and benefits and payers cannot accurately predict the utilisation of health care services.”

A distorted picture

There are a number of ways in which trial reports paint a distorted picture: patient selection for trials, a failure to detect or report side effects, and the way data are presented are all implicated.

Patient selection

Patients who are fit enough to join clinical trials are not representative of the substantial proportion of patients with the condition in the wider public. Trials usually exclude those with heart or kidney disease or a previous history of cancer. It has been half-seriously suggested that to enter a clinical trial you need to be “a marathon runner who happens to have cancer”.
This means that when drugs are used in clinical practice, results very often don’t live up to expectations. Niraula says, “Drug companies put a lot of investment into clinical trials, and mostly with good intentions want the drug to work for the benefit of the patient and understandably, want a return on their investment. When it enters the real population, the result is a higher likelihood of toxicities and a lower likelihood of benefits.”

A study at the Princess Margaret Hos-pital in Toronto provides some confirmatory evidence. It compared outcomes for patients with metastatic castration-resistant prostate cancer, treated at the same hospital, to identical standards of care, according to whether or not they were on a trial. They found that the trial patients were younger, had less comorbidity and better performance status. Patients treated in routine practice had shorter survival and experienced more toxicity, notably fever and infection (Ann Oncol 2013, 24:2972–77). This difference between outcomes inside and outside clinical trials even has its own label: “the efficacy–effectiveness gap”.

The likelihood is that differences in outcomes will be even greater for patients treated away from major centres, since patients are likely to have poorer access to supportive care to address side effects. As quality of life worsens, patients may suspend treatment or reduce the recommended dose.

Age discrepancy is widespread within clinical trials, as a by-product of excluding patients with comorbidities. The CML Advocates Network found that the average age of CML patients on phase III trials was 47, while the average age of real world patients in Europe is nearer 65, meaning that side effects in the older population with comorbidities are not discovered in trials.

There is, however, evidence that exclusions do not necessarily invalidate trial results. In a study with some similarities to that conducted in Toronto, Joseph Unger and a team at the Fred Hutchinson Memorial Hospital in Seattle studied 21 RCTs supported by the National Cancer Institute. By comparing the survival of patients on the control arm – who were receiving standard care – to similar patients treated outside trials, they were able to gain insight into differences relevant to being in a trial (JNCI 2014, 106:dju002 doi:10.1093/jnci/dju002).

Unger and his colleagues found that, while being on a trial was associated with better survival, the difference lasted for only one year after diagnosis. They believe the difference is simply due to patients in the trial being younger and fitter with fewer comorbidities.

Survival curves for standard treat-ment patients in trials and non-trial patients were very similar in the longer term. Of course, looking at control arm patients does not say anything about the efficacy of treatments, but Unger says it suggests that any benefits found for new treatments should translate to a real-world setting. “The fact that over the long term patients had very similar outcomes suggests that trials are not pick-ing off qualitatively different cancer patients, they are just excluding those with comorbid conditions that affect survival in the short term.” However, this conclusion would not be valid, he says, if the treatments have too much toxicity or poor compliance.

Unger is also concerned that some of the exclusions of patients due to concern over safety are outdated, and that it is increasingly unrealistic to exclude patients from trials simply because they have had a previous cancer.

However, the tendency to exclude patients appears to be increasing. A study of 86 practice-changing RCTs showed that the proportion of patients excluded from trials had doubled to 18% after 2010, compared with the 9% before 2000 (Cancer Treat Rev 2016, 43:67–73). There were increases in exclusions of patients with cerebrovascular events, gastrointestinal bleeding or cardiac conditions. There was also a decrease in the average upper age limit.

the price...

The price we pay for progress A 2012 study of 12 widely used targeted cancer drugs approved since 2000 (JCO 30: 3012–19) showed that most are associated with higher rates of toxic death (odds ratio1.4), treatment discontinuation (OR 1.33) or severe adverse events (OR 1.52)

Trials in denial

Deciding on which side effects to look for can influence what is found. Ian Tannock, a leading member of the Toronto group, believes the ATAC trial led to a distorted view of the relative safety of Arimidex to tamoxifen, because it was left to the doctors involved in the trial to make a judgement on which events could be connected with the treatment. In a letter to The Lancet Oncology (March 2011), he argues that this created a bias “because side-effects due to tamoxifen were recognised better at the start of the ATAC trial than were those due to anastrozole,” and he suggests it would be better to have a prespecified checklist.
Patients, however, point out that prespecified checklists can also lead to under/non-reporting of important side effects that have been omitted from the list. This is a particular problem for side effects such as exhaustion and diarrhoea, which are not life-threatening but can make life almost unbearable.

Gilly Spurrier-Bernard, president administrator of MelanomaFrance, describes how difficult it was for her husband to record side effects on a trial of vemurafenib (Zelboraf), despite being under the care of the Gustave Roussy Institute, one of Europe’s best cancer centres.

“Clinicians only want to report the effects that the trial pharmaceutical companies have identified as a high risk. My husband had a number of skin reactions which we knew were to do with the drugs, because he had never had them before, and they look down the list and say, that is nothing to do with the trial.

“We were treated at a very good centre but it used to drive me up the wall that what you were reporting as potential side effects did not even get recorded.”
Several studies show that clinicians under-report adverse events that are very significant for patients.
In 2015 a study from the Italian National Cancer Institute in Milan found extensive under-reporting by doctors of six symptoms that blight the lives of patients in three randomised trials, including nausea, diarrhoea and anorexia (JCO 2015, 33:910–915). Six years previously, in 2009, a survey by Myeloma Patients Europe had shown fundamental differences in perceptions between patients, nurses and doctors in assessing the impact on quality of life of various side effects, including hair loss, fatigue, reduced body function, neuropathy and thrombotic events.

Eric Low, chief executive of Myeloma UK and the chief author of that report, says it shows why patients must have more of an input into reporting side effects to ensure that trial reports paint an accurate picture. He points out, however, that it is only when drugs come into everyday use that clinicians learn how to deal with side effects. He gives the example of bortezomib (Velcade), the first significant proteasome inhibitor, which was given accelerated approval in 2003 as a treatment for relapsing myeloma.
“Initially bortezomib had many side effects, particularly neuropathy, but over time we got a subcutaneous version and doctors moved to giving it once a week and that made a dramatic difference. Now peripheral neuropathy is quite rare. “The real benefit of a new drug comes as clinical experience accrues and patient management and patient selection improves. At the point where a new drug is approved we don’t have in depth data, and with a move towards accelerated approval we are going to have even less.”

Data from general clinical practice is, however, only used to update clinical trial reports in a small minority of cases. Bostjan Seruga reported that his team had looked at 311 RCTs of prostate, breast and lung cases published over a 30 year period and found that only one in five had published updated reports. Where publications were updated they predominantly showed a smaller magnitude of effect and a 
greater number of side effects, than the original reports.

There is increasing support, by EMA in Europe and the NCI and the FDA in the US, for moving towards patient-reported outcomes to mitigate the inaccurate reporting of side effects. The EMA completed a public consultation on this issue in 2015 and is expected to report back early this year.

The issue is complicated by the fact that, in the context of certain clinical trials, patients themselves may feel they have an incentive to downplay the seriousness of side effects. Gilly Spurrier-Bernard knows this from her own family experience, when her husband was on a trial for ipilimumab, and in her advocacy role hosting online forums for melanoma patients.
“I spent four years filling in patient questionnaires and as far as I am concerned they are totally useless. Patients lie through their teeth because they know that patients get kicked off the trial if they show any slightly scary signs of side effects. With the ipilimumab trial the slightest sign of colitis or diarrhoea of significant amount you were pretty much kicked off. This is all discussed on patient forums.”

She fears for what will happen when the trial treatments come into general use. “People with brain mets, or comorbidities or lupus are excluded from most of these trials. How will side effects affect people who already have autoimmune problems? None of this has been recorded properly. They need to get it sorted.”

Misreporting data

Whether by accident or by design, the process of writing up clinical trials offers further opportunities to downplay the negatives and talk up the positives.
In 2004 An-Wen Chan and colleagues reported on 122 journal articles from 102 clinical trials and found that 50% of efficacy outcomes and 65% of harm outcomes were incompletely reported (JAMA 2004, 291:2457–65). In 62% of trials, at least one primary outcome from the trial protocol was changed or omitted. The authors concluded that “reporting of trial outcomes is not only frequently incomplete but also biased,” and that “published articles may overestimate the benefits of an intervention.”
Another of the landmark studies from the Princess Margaret Hospital, Toronto, found that a third of clinical trials for women with breast cancer showed “bias in reporting” in primary endpoints, and two thirds showed bias in reporting tox-icity (Ann Oncol 2013, 24:1238–44). Positive trials were particularly associated with under-reporting toxicity.
Peter Jüni, Founding Director of the Clinical Trials Unit of Bern University Hospital, outlined at the 2015 European Cancer Congress how the reported results of clinical trials are often distorted. Common practices include ‘fishing’ through data for spurious positive outcomes, swapping primary and secondary outcomes because the primary outcomes are not very good, and excluding outliers to make results statistically significant.
Perhaps the most pernicious practice is selectively omitting inconvenient results, such as the 2% of patients on the cetuximab arm who died as a result of cardiopulmonary arrest. A bigger problem may be the non-publication of entire trials that generate inconvenient results. It is such practices that sparked the launch of the AllTrials campaign in January 2013, which calls for “all trials past and present [to be] registered, and the full methods and the results reported” (alltrials.net).

Incomplete information

StampaLaws governing the marketing of medicinal products in European Union member states require that all medicinal products must “be accompanied by labelling and package leaflet which provide a set of comprehensible information enabling the use of the medicinal product safely and appropriately”.
But research into the reporting of side effects for some of the most widely used targeted anti-cancer drugs shows the majority are not reported in the pivotal trial and are added to the label, sometimes many years later.
The TKI HER2-blocker lapatinib (Tykerb) has had 12 amendments to its label since it received marketing approval in March 2007, even though safety had already been evaluated in clinical trials in more than 3,500 patients with advanced or metastatic breast cancer. The most common adverse reactions (i.e. in more than 20% of patients) initially recorded for Tykerb plus capecitabine were diarrhoea, hand-foot syndrome, nausea, rash, vomiting, and fatigue.

Warnings given on the label included:

  • Reports of decreases in left ventricular ejection fraction
  • Foetal harm if administered during pregnancy
  • Dose reduction to be considered for patients with severe hepatic impairment.
  • Prolonged QT interval in the heart’s electrical cycle in some patients.

In August 2007 further warnings were added about:

  • Interstitial lung disease and
  • Pneumonitis

In 2008 a boxed warning (highest grade of warning) was added about:

  • Reports of severe and sometimes fatal hepatotoxicity – “If changes in liver function are severe, therapy … should be discontinued”

Various notices were added about drug-drug and drug-food interactions in the intervening period.
In June 2013 the label was amended to warn 
about:

  • Grade 3/4 diarrhoea. “The diarrhea may be severe, and deaths have been reported,” says the label. (Most cases of diarrhoea are less severe, occur 
early in treatment and last 4 to 5 days.)

Wrong dosage, worse effects

While many of the biases listed above may be nothing new, it seems that reporting of side effects from targeted drugs may be a particular problem. One reason is that cytotoxics are prescribed for fixed protocols, whereas targeted drugs are often continued until resistance develops, and adverse effects that are not immediately apparent often occur later.
The big problem here is not just that, as Seruga remarked, it is the early impression about lack of clinical harm that sticks, but that early toxicity results set the basis for defining dosing, and as a result recommended dosage levels may be set too high.
Research led by Sophie Postel-Vinay from the Gustave Roussy Institute found that more than half of the most serious toxicities in phase I trials occurred after the end of the ‘dose-limiting-toxicity’ period used to determine tolerability (JCO 2011, 29:1728–35). Although the severity of toxicities decreased during the trials, the proportion of unresolved toxicities increased, more medication had to be prescribed to deal with side effects, and dose reduction became more frequent, suggesting that “benign late toxicities may not be bearable over time and might require specific management.”
This was confirmed in a much larger study led by Postel-Vinay and coordinated by the EORTC, which gained unprecedented access to raw patient data from institutions and pharmaceutical companies covering more than 2,000 patients in 54 phase I trials (EJC 2014, 50:2040–49). Almost half of patients who suffered severe side effects (grade 3 or worse) had their first episode after the cycle of treatment that was used to define dosage. One in 11 patients experienced dose-limiting side effects (i.e. the medication had to be paused or reduced), of which the most common were fatigue, nausea, vomiting, gastro-intestinal disorders and hypertension.

The way forward

Most experts agree on a number of steps to improve reporting on data from clinical trials and assess the value of new drugs.
Saroj Niraula, in Winnipeg, says that good-quality population-based studies are required from real-world use after a drug receives full approval, along with stricter regulations about reporting. “We as physicians should be able to provide our patients with the most comprehensive information possible on efficacy and toxicity before they come to a decision about the amount of toxicity that is acceptable to them for a given benefit.
“Journals have to be more stringent. There should be academic incentives to report toxicities well. We want honest and exhaustive information from pivotal drug trials.”
Joseph Unger at the Fred Hutchinson in Seattle believes that trials should have fewer exclusions. “From a patient perspective access to trials is a huge issue. But also from a researcher’s perspective we want to be able to do these trials as quickly as possible. If we are excluding patients for reasons that are unneces-sary, that is hindering our efforts.”
At the Gustave Roussy, Sophie Postel-Vinay is calling for data on adverse effects to be collected more comprehensively and for longer periods. “The key recommendations are that everything about late toxicities is reported, which is not the case at the moment, and that the recommended phase II dosage is based on everything that is seen over the whole trial.”
These recommendations are already being adopted in protocols or written into guidelines for some phase I trials, although there is as yet no settled methodology for deciding on the dose limiting toxicity definition and duration, or the phase II dose recommendation.
Gilly Spurrier-Bernard from MelanomaFrance is campaigning for a patient-driven reporting system filled in on laptops or phones whenever there is a significant event, as some patients already do with pain diaries. “Patient issues change over time and according to how healthy you are feeling. Researchers need to be asking how it impacts on daily life. Then you need some clever algorithms for data mining.”
Bettina Ryll, who founded Melanoma Patient Network Europe after her husband Peter developed malignant melanoma, agrees. “We see more and more selected trial populations and it automatically becomes less representative of the entire patient population,” she says. “RCTs are the wrong way to tackle safety. We need a much better pharmacovigilance system where we capture data much more systematically and then act upon it.
“We need new drugs, as every patient with a life-threatening condition will tell you. We also need a way to study them meaningfully and in a way that does not prevent access for patients, does not drive up cost and captures reality.”
Melanoma Patient Network Europe is preparing a project with the Uppsala Monitoring Centre to harvest direct patient reports of symptoms and side effects. The Centre runs the WHO international drug monitoring programme, which was set up after the thalidomide disaster, and has the world’s largest dataset of adverse events, publishing data from 120 national health authorities worldwide on an open website at 
vigiaccess.com.
Bostjan Seruga from Ljubljana would like to see the American NCI initiative on patient-reported adverse events (PRO-CTCAE) fully incorporated into clinical trials, along with updated reports to capture data not originally reported by RCTs, and specific trials to address the needs of patients who were ineligible.
“Oncologists, journal editors and societies like ESMO and ASCO need to introduce measures to ensure complete reporting of toxicity to serve our patients better.”

Goodbye or arrivederci?

Our new vocabulary – gene, genome, molecular, targeted, personalised – no longer inspires the confidence it once did, and as the cancer community prepares to gather at the 18th ECCO – 40th ESMO European Cancer Congress in Vienna, there is a real sense of uncertainty about where the next major progress will come from, writes Alberto Costa in this editorial.

Where will European oncology go from here? This is a question many of us will be asking as the ECCO–ESMO congress convenes in Vienna. There’s a diffuse sense of uncertainty, coming mainly from the laboratories, where many promising cutting edge innovations still seem to be in the air. Our new vocabulary – gene, genome, molecular, targeted, personalised – has lost its novelty and its shine. What will be the next clinical trial to have us all breathlessly awaiting the results? What innovation will be the next to radically change our clinical practice? A second generation of Da Vinci robots for everybody? Intraoperative radiotherapy? Immuno-oncology? Alopecia preventing devices?

With this in mind, the thoughtful ECCO–ESMO participant will also be worrying about the endless list of cost issues that intrude on clinical decisions. This is not something we were prepared for; we never studied pharmaco-economics (or device-economics) at medical school. How can clinical oncologists take these decisions? Is what we do even still clinical oncology, or is it a highly complex combination of medicine, nursing, ethics, sociology, economics and politics?

On top of this, many of our friends participating in ECCO–ESMO will want to attend sessions that address questions about how and where care should be delivered to their patients. Questions like: should I send all my breast and prostate cancer patients to the nearest certified breast or prostate unit? It’s now accepted that all patients with rare cancers must be referred to the nearest centre of excellence, but what about other patients? Can I, a surgical oncologist, continue to practice as I have done for the last 20 years? Is it still OK to ’do’ a lung cancer one morning and a liver cancer the next? Can I, a medical oncologist, safely treat a patient with an advanced colorectal cancer, a bone sarcoma and maybe a lymphoma, all within the same outpatient clinic?

These are our common concerns and the things that really matter to all of us who are proud to attend the ECCO–ESMO conference. The Americans have decided to keep well separated the physician researchers (AACR), the cancer doctors (ASCO), the nurses (ONS) and the patient advocates. Here in Europe we have a long tradition of working together, but the will to continue to do so is now in danger.

The details of how ECCO and ESMO should collaborate may be of no great interest to participants at the Vienna conference, but the great majority will undoubtedly feel that staying together is the right thing to do, both for cancer health professionals and patients. Cancer has become all about collaboration, and it’s too late for any single specialty to work in isolation.

When we leave Vienna, we want it to be with an arrivederci and not goodbye.

Asking the dumb questions

It took ten years of immersion in the world of cancer research to produce the book that won Clifton Leaf a Best Cancer Reporter Lifetime Achievement award. But it started from a simple question: how do claims about winning the war on cancer square with a failure to cut death rates?

When Clifton Leaf was working as the Wall Street editor at Fortune magazine in the early years of the new millennium, it seemed there was no shortage of scandals to cover – from the accounting crimes of auditor Arthur Andersen to the house of cards that was Enron. At their heart was a fundamental driver, Leaf said: “Greed. Many of the central figures in these Wall Street scandals were rich already – it was incredible to see that some would do anything to get even richer.”

However, when it comes to the world of cancer research and treatment, where Leaf has also spent a decade saying that the numbers do not add up, he has nothing but kind words about the people.

“When I got into the cancer enterprise, I got to talk to lots of people at the beginning, and that now runs into thousands. I did not meet anybody who would not have given their right arm to have cured cancer. There was a different passion and drive and a willingness to engage in the problem that I had never experienced.”

This might sound odd, given the storm Leaf stirred up about the failures of cancer research and treatment, and the way the ‘war on cancer’ has been mishandled. But he has always been clear about the difference between the policies and the people, and he learnt about life’s many contradictions early on. At the age of 15 he was diagnosed with advanced Hodgkin disease and experienced a combination of drugs and radiation therapy that almost killed him even as it saved his life.

Now Deputy Editor of Fortune, Leaf recently went to Switzerland to receive a Best Cancer Reporter Award for Life-time Achievement from the European School of Oncology. His work stretches from his 2004 cover story, ‘Why we’re losing the war on cancer – and how to win it’ to his 2013 bestselling book that elaborated on reasons for this failure, The Truth in Small Doses. Among many articles in between was his 2013 cover story for the Sunday Review section of the New York Times, ‘Do clinical trials work?’

Over this decade Leaf has succeeded in reframing the story of cancer. The usual narrative has science relentlessly unveiling the secrets of this terrible scourge and developing ever more brilliant drugs to treat it. Indeed, when he first came to the topic this is what Leaf believed. “My bias was that of the average American. You only know what you read in the paper. I thought we were getting one breakthrough after another.”

He came with no science background, having slept through most of the biology course at high school and never having written about medical science. However, this was a time when Glivec, Avastin and Erbitux were being hailed as wonder drugs, and there was immense interest in the field from investors. As a business reporter he could not ignore it.

“I started thinking about the cancer problem as if it were a corporate enterprise. Instead of a profit and loss statement or a balance sheet, I asked, ‘What are the metrics that show how the cancer enterprise is doing? How many people were getting the disease each year, how many were dying and how much were we spending?’ The answers were neither clear-cut nor encouraging. Those numbers seemed to be going the way of Enron, and I started to wonder if things were as they should be.”

His main conclusion was that you cannot claim a success so long as the cancer burden – the overall number of people developing cancer and dying from the disease – continues to rise. The aging population is a critical factor, Leaf acknowledges. But he says we have to address these population demographics head-on, work to lower the number of people getting cancer and focus on pre-empting the disease process earlier in its progression.

In the US alone, about 230,000 women will be diagnosed with invasive breast cancer this year and another 60,000 with in situ disease. “My question is, how is that winning? We can’t say we have a victory because we say we have slightly reduced the age-adjusted death rate.

“Even when they make it through five or six years of treatment many of them still die. They are counted as successes because they exceeded the five-year survival rate but are no longer with us. Since I began this enquiry ten years ago I have collected many people in my life who reached out to me or I met at conferences. I have lost many of them along the way. You hear about it in night-time calls, e-mails or text messages – it is brutal.

“I measure progress by whether the burden of cancer is being reduced or not. I measure the burden by the number of people going through this gruelling awful process.”

The dumb questions

The last time Cancer World wrote about Clifton Leaf (2007) the headline described him as “asking the difficult questions”. He demurs. “I think they are the dumb questions – the really straightforward stuff. Like: at what point does making sure that we have fewer women getting breast cancer become as paramount as treating it?”

His book focuses on the mismatch between the brilliance of the scientists and what he sees as the minimalist ambitions of the clinical trials enterprise – citing Irv Krakoff on research “aiming to find significant answers to insignificant questions.” He describes how the US war chest for cancer goes to the same institutions year after year, how researchers spend 50% of their time applying for grants and filling in paperwork, and how young researchers with bright ideas spend an increasing amount of time doing non-inventive experimental work in large laboratories, gathering an endless amount of ‘preliminary data’, while their ambitions and inspiration wither.

He details the lack of progress on finding cancer markers for early diagnosis and the minimal sums spent on prevention. Meanwhile, as the death toll continues to rise, every step for¬ward is hailed as a breakthrough, especially on the business pages. “Company stocks go soaring on merely the wisp of good news from clinical trials and they fall precipitously when something goes awry.”

Part of the problems is the filtration process. “Companies present their data in lofty conferences and the world gathers with bated breath as if a new pope was being chosen. The low expectations that set the context for clinical trials help shape these dramatic responses. If you are used to the fact that nothing works, and something comes along that improves survival by two months, there are hosannas and angels singing and the stock goes through the stratosphere.”

There is much more of this in the book, which is fast paced as you might expect from a journalist. He focuses on people who challenge the status quo, and are often marginalised until they turn out to be right.

Undoubtedly his work has had an impact, but Leaf does not consider himself an expert. “I would definitely say expert is the wrong word. I would say I am a hard-working remedial student, well-studied in the way that a remedial student needs to at least be able to communicate with the people I am having a conversation with.”

This is one reason that his book includes 81 pages of (often chatty) endnotes and almost 100 pages of references at the back. “I made a real effort to make the sources and references as comprehensive and readable as I could, and used the top-tier journals. I was holding up a mirror and saying, ‘this is what you in the medical and scientific community have discovered through your hard work and training’. This is not me saying this. All I am is an aggregator or scribe.”

He admits to being a good polemicist. “But what I really wanted to be was a story teller and to have a sense of a shared conversation. I did not think people would listen closely to a long argument any more than they would listen to a bore at a cocktail party.”

His writing is not just about what has gone wrong. He says that finding good solutions requires good management and good engineering. As deputy editor of Fortune magazine, he is responsible for seeing the print edition to press and coordinating the work to gel at the right time. “We get close to missing the deadline every issue but never do. We make it time after time only because it is managed.” He says that levels of creativity and innovation and new ways of thinking about story telling are encouraged, not discouraged, by the process.

However, he accepts that many scientists fear it. “There is this idea that if you are being managed there is no way you can have independent thought or creativity or innovation.”

Ask Google

He suggests that cancer researchers learn from the business world. “If you look at Apple or Intel or Google or Facebook, so many explosive ideas are possible because of brilliant manage¬ment. The aim of management is to free up the creative idea and to shape and facilitate innovation. What is happening in the cancer world, frankly, is also management – researchers and clinicians are being micromanaged and mismanaged to exhaustion. Sometimes all ‘good management’ entails is putting an end to the micromanaged systems that don’t work and shaking up a culture of deadly caution.”

He calls for smaller but longer term grants that free the most creative scientists to get on with their research and a better ‘sys-tems approach’ to organise trials that we can learn from more quickly and reduce ‘me too’-ism. He cites the hopeless coordination in the global hunt for cancer biomarkers and contrasts it with the best business approach. Maybe progress can be made by learning from the likes of Google, a company that encourages hundreds of pieces of innovation in its labs and then throws mass resources at developing those that look most promising.

“There has to be some process to choose what is worth pursuing and what is not and how much effort to put in and where the resources come from and that needs focus. I think of engi¬neering as bridge building, making sure that people and materials are there at the right time.”

Leaf does see some progress over the decade he has been beating this drum. There is greater recognition that young scientists need to be given more backing at a time when they are most creative. But he sees little progress in the way that the big grants are doled out in the USA – a bureaucratic and risk-averse process that he believes stifles innovation and enforces conformity.

One big area of change has been the emergence of a new breed of patient advocates. “They are not just here to raise money and wear pink ribbons and march, but to help solve problems. That means helping to recruit for and shape clinical trials; making sure that the questions being asked are the important ones; that the right markers are being used to stratify the right patients; that trials are appropriately controlled. They are engaging more with institutions.”

Until the number of people developing cancer and dying from it starts to fall, Leaf does not feel that anyone can claim to be winning a war against cancer. So will he continue to be an active voice?

Leaf has a demanding job and a family for whom he wants to be present and involved. He does not have the time for research and writing in the way that he did, but does not intend to disappear. He is a regular keynote speaker at conferences and is often invited to sit on panels and boards, including three times on the President’s Cancer Panel Meeting.

“I don’t think you can open up your mouth as loudly and widely as I did and then just walk away. It is kind of cowardly to throw stones and not wait for everyone to come and confront you.” In fact he has become friends with many people in cancer research who disagree with him and he sees it as a healthy sign that they seem to enjoy being challenged (as he does).

“I have been lucky enough to have a platform, first with Fortune magazine and then through the brilliance of the cancer community at large, then the book with Simon & Schuster. Because I have had that benefit I am going to stick around for a while and see if I can be of help in a collaborative way of keeping the conversation going. I think that is important.”

When interviewed for Cancer World in 2007 Leaf observed that many of his critics were predicting that the big cancer breakthroughs would come by 2015. A quick look at the calendar would suggest that, unlike his magazine, his critics have missed a deadline.