Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts by MaverickNY

Yesterday was a travel day but thanks to gippy wifi and a packed day, I didn't have the opportunity to post about some interesting articles on prostate cancer published in the NEJM, which I read and digested on the train up to Boston. Now that the embargo has lifted, we can all talk and write about the data publicly.

Kantoff et al., have reported the full results of the IMPACT trial data for sipuelucel-T (Provenge) from Dendreon (a client), which led to the vaccine being approved by the FDA for the treatment of asymptomatic prostate cancer recently.   The results are not new and contain no surprises as they are already well represented in the prescribing information (PI), which are available online for anyone to read and have been presented ad nauseum at recent meetings at the American Urology Association (AUA) and American Society of Clinical Oncology (ASCO).

For those who missed the meetings, the data can be summarised by the following quotes from the article:

"This reduction represented a 4.1-month improvement in median survival (25.8 months in the sipuleucel-T group vs. 21.7 months in the placebo group." 

"The 36-month survival probability was 31.7% in the sipuleucel-T group versus 23.0% in the placebo group." 

"Adverse events that were more frequently reported in the sipuleucel-T group than in the placebo group included chills, fever, and headache."

What can conclude from this is that the vaccine helped men with asymptomatic prostate cancer live longer without the known significant toxicities seen with chemotherapy such as alopecia (hair loss), severe myelosuppression, neutropenias, anemias, febrile netropenias and infectious complications. That would be a win in most people's books because people want to know whether their relative with cancer will live longer, feel better than they do and have a reasonable quality of life with their families. The price might be a bit of a sticker shock, but insurers seem to be covering it to date without any major hiccups.

The accompanying editorial, however, from Dan Longo, a cancer researcher and Scientific Director, National Institute on Aging, National Institutes of Health in Maryland, was probably well meaning, but rather surprising and a little harsh at times.

Science is about the pursuit of truth and in this case, finding out whether men with an advanced cancer would live longer, which they clearly did, so to grumble about the lack of tumour shrinkage is a little odd. Oncologists have for years focused on this as an indication of response rate through a measurement system known as RECIST although whether the patient lived any longer was somewhat moot.  In Longo's editorial, he noted:

"It is hard to understand how the natural history of a cancer can be affected without some apparent measurable change in the tumor, either evidence of tumor shrinkage or at least disease stabilization reflected in a delay in tumor progression."

This is rather old school thinking and as we develop new therapeutic approaches, the end points and surrogate biomarkers may well change.

There have been far too many studies in cancer that showed that traditional chemotherapy could shrink a cancer (great) while toxically poisoning everything in its wake as well (not so great), making many of us wonder whether this was actually a useful approach if survival wasn't impacted. By this, I mean are we measuring the right thing in the first place? 

In full disclosure, my own Father passed away from this disease 10 years ago this month, so it possible that I'm biased in my thinking have gone through his experience with him.  It was a very stressful time for all of us, him especially, although he held up well and maintained his dignity until the end.  Still, I think I would rather know that a new therapy would help a male relative of mine lived longer (improved overall survival) and felt better (less sickness and pain). Then we could all enjoy their last few years together in dignity with quality time than go through the hell that is chemotherapy, which he chose not to do because after many discussions, because if it wasn't going to cure him he saw no point in putting my Mother through that agony.

Sometimes, we need a more humanistic approach to oncology and to remember that these are people's lives we are talking about, not a number on a stat sheet. Tumour shrinkage?  "Pshaw, what does that mean?!" was my Dad's cynical reaction.  Looking back, he was instinctively right.

Another question that was raised in the editorial was the study design, ie was the control group an ideal one? Well, to be fair the time for input into those issues is during the protocol design stage when many people's input is sought before a trial even begins enrolling. Grouching about it after the event seems a little churlish at best. 

Could the control group have experienced some stimulatory effect?  Possibly, but then when you look at the data, the survival time is not out of kilter with those from other control groups previously reported versus chemotherapy who mostly received best supportive care, ie usually pain medications and growth factor support. Perhaps had that approach been taken the difference between the control group and the sipuleucel-T group would have been even greater.

"As the authors point out, differences in subsequent treatments (e.g., docetaxel) do not appear to account for the survival differences, but methods for assessing such effects are imperfect. New prognostic variables such as statin use, the duration of the first off-treatment interval, circulating tumor cells (as assessed as EpCAM+CK+CD45− objects), and new prognostic algorithms may need to be accounted for in assessing therapeutic effects."

Well quite, but I'm sure we'll hear more about further meta analyses of the data at some point. There are a number of other vaccines in phase II/III trials and as we learn more about how this therapeutic approach works, which markers to measure and what impact they have on both the immune system and the natural course of the disease, so our scientific and clinical knowledge will improve and hopefully, patient outcomes as well.

Promising phase II data doesn't always translate into significant phase III data, as we have seen more compounds and vaccines than I remember over the years. For now, we have two new therapy options for men with prostate cancer at different stages of the disease, more choice can only be a good thing, as is having a active pipeline for treatment of prostate cancer, offering hope for many that progress is being made after years of stagnation.

{UPDATE: I spotted Adam Feuerstein's (The Street) and Matthew Herper's (Forbes) excellent articles on this topic after writing this blog post, so they are linked for further information and background reading for those interested.

ResearchBlogging.org

Kantoff, P., Higano, C., Shore, N., Berger, E., Small, E., Penson, D., Redfern, C., Ferrari, A., Dreicer, R., Sims, R., Xu, Y., Frohlich, M., & Schellhammer, P. (2010). Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer New England Journal of Medicine, 363 (5), 411-422 DOI: 10.1056/NEJMoa1001294 

Longo, D. (2010). New Therapies for Castration-Resistant Prostate Cancer New England Journal of Medicine, 363 (5), 479-481 DOI: 10.1056/NEJMe1006300

A while back I wrote about how circulating tumour cells (CTC’s) see here and here, can be used as a potential new surrogate measure for prostate cancer, so it was with great interest that I read an excellent article (free public link courtesy of AACR) on abnormal circulating cells in non-small cell lung cancer (NSCLC) late last week.

Essentially, this new research from three academic institutions (MD Anderson TX, Manchester UK and Baltimore, MD) set out to:

“Determine if a fluorescence in situ hybridization (FISH)–based assay using isolated peripheral blood mononuclear cells (PBMCs) with DNA probes targeting specific sites on chromosomes known to have abnormalities in non–small cell lung cancer (NSCLC) cases could detect circulating genetically abnormal cells (CACs).”

In the recent discussion about fluid-based blood biomarkers, we saw how people could be spared invasive and inconvenient tumour biopsies, which are not always practical in advanced lung cancer, particularly if the patient is elderly or frail.  In this study, the researchers demonstrated that detection of circulating epithelial cells (CECs) or CTCs using a simple blood test may assist in early detection of lung cancer at diagnosis and relapse and provide a minimally invasive way to monitor results of therapy.

Now in this study, only a small number of people with NSCLC were evaluated (NSCLC n=59 plus n=29 controls), but they do give us an indication of what could be looked at in larger scale clinical trials (first find your needle in the haystack!)  The presence or increased numbers of these circulating abnormal cells confers a poorer prognosis, ie associated with relapse of disease and poorer survival, so the ability to pick them up earlier and more easily would be an advantage clinically.

The approach used in this research was a commonly used technique called fluorescence in situ hybridization (FISH) to detect abnormal circulating cells that have aberrations found in non-small cell lung cancer. FISH detects and quantifies abnormal cells by using dye-labeled DNA probes of cell chromosomes that cause cells with the targeted genetic abnormalities to light up when viewed under a fluorescent microscope.

The researchers chose 12 biomarker probes that target aberrations previously connected to lung cancer to analyze both the controls (people without lung cancer) and people with NSCLC, including both smokers and non-smokers.  In the analysis they found the following:

  • Highly significant differences in the average number of abnormal cells in the bloodstream between patients and controls.
  • Abnormal cells were significantly associated with disease stage, with cells that contained certain abnormalities increasing significantly as cancer progressed from early to advanced stage disease.
  • Eight of the biomarkers had a strong overall correlation between abnormal circulating cells and tumors.  Chromosomal gain of the EGFR gene in circulating cells was significantly associated with the same gain in tumors, particularly in patients with stage III or stage IV disease.

No doubt work will soon be underway to develop a commercial clinical test based on FISH, which would then be able to be used in the Community Oncology setting, where the majority of patients are treated in the US.  Validation in clinical trials will be crucial to this process.

In an MD Anderson news release, the lead author, Ruth Katz, was quoted as saying:

“Blood tests for these circulating tumor cells could be used to diagnose lung cancer earlier, monitor response to therapy and detect residual disease in patients after treatment.”

Source: MD Anderson Cancer Center

Having an easier to use test to monitor responses more accurately to products in development will be particularly useful for pipeline drugs.

References:

ResearchBlogging.org Katz, R., He, W., Khanna, A., Fernandez, R., Zaidi, T., Krebs, M., Caraway, N., Zhang, H., Jiang, F., Spitz, M., Blowers, D., Jimenez, C., Mehran, R., Swisher, S., Roth, J., Morris, J., Etzel, C., & El-Zein, R. (2010). Genetically Abnormal Circulating Cells in Lung Cancer Patients: An Antigen-Independent Fluorescence In situ Hybridization-Based Case-Control Study Clinical Cancer Research, 16 (15), 3976-3987 DOI: 10.1158/1078-0432.CCR-09-3358

Our lives are filled with the shadows of stories unwritten. Our lives are filled with waiting and hoping and putting off and stuffing the cracks with papers from other people’s lives.

If you KNEW without a doubt that your decisions were the measure of who you are – and make no doubt that they are – are you PROUD of your decisions? Are you living the story you want to live?

And what will you do with tomorrow that you haven’t done with today? What lighthouse will you point yourself towards? And what flag will you fly?

Live now. Don’t wait. And fill in some of those shadows with stories you’d want to see written about you.

Chris Brogan’s guest post on @julien’s blog today really made me stop, think and wonder.

How many of you are out there waiting for something to happen?

Think of the drug pipelines to move, the new indications to drive… Are you moving them forward purposefully or sitting back in the dog days of summer?

This is our busiest time of the year, when catching a moment to take a breather and reflect is harder, but judging by our clients some folk are out there pushing things along and trying to make a difference to the lives of people with various diseases.

Carpe Diem.

Posted via email from sally church’s posterous

We all know that long term exposure to chemical insults and carcinogens from smoking and alcohol can act as irritants, leading to biochemical changes that can induce the development of hyperplasia, and potentially, carcinomas.

Over the weekend I was shocked to learn that Alex "Hurricane" Higgins, former World Snooker Champion, had passed away aged 61 from throat cancer.  He was an incredible talent who will be sadly missed and remembered for many years to come.

I wrote a bit more about it here: be warned though, the before and after photos are rather distressing 🙁

2 Comments

We've heard a lot about the impact of KRAS in colorectal cancer as a useful biomarker for determining whether or not to treat with EGFR therapy, depending on whether the mutation is wild-type or mutated, but now new evidence has emerged for it's possible role in ovarian cancer.

A paper by Ratner et al., from Yale University, looked at the relationship between an increased risk of OC and a KRAS variant.

Overall, the data demonstrated that a variant of the KRAS oncogene was present in 25% of all ovarian cancer patients. The same variant was also found in 61% of ovarian cancer patients with a family history of breast and ovarian cancer. Up until now though, it has proven impossible to tell which women with a strong family history of breast and ovarian cancers would go on to develop the disease. These results therefore offer some new clues in the puzzle.

Related research from another team from the UK, Denmark and US, published last year, looked at tagging single-nucleotide polymorphisms (SNPs) in candidate oncogenes and the susceptibility to ovarian cancer in approx. 1,800 women with invasive ovarian cancer compared with controls (n=3,000). The goal was to identify moderate/low-risk susceptibility alleles of the proto-oncogenes BRAF, ERBB2, KRAS, NMI, and PIK3CA, but no evidence of ovarian cancer association was found with these SNPs. When stratified by
histologic subtype, however, one common variant allele have borderline evidence of association with epithelial ovarian cancer.

What was interesting to me in the latest research from Yale is that all the women in the study had a strong family history of cancer, but only half had known genetic markers of ovarian cancer risk such as BRCA1 or BRCA2 mutations. The results, albeit from a small sample (n=157), suggest that the KRAS variant may offer a more sensitive than currently available. 

The Yale group had previously established that the KRAS-variant is not somatic but germline, meaning it is identical in person's normal and tumour tissues, thereby enabling the researchers to collect primarily germline DNA from either blood or saliva, rather than from tumour biopsy samples.  This is huge from a patient perspective and physician point of view in terms of ease of use and convenience.

This study is important because ovarian cancer is the single most deadly form of women's cancer and is usually diagnosed in advanced stage disease.  Part of the reason is because of the lack of known risk factors or genetic markers of risk.  To this end, the authors concluded:

"Our findings strongly support the hypothesis that the KRAS-variant is a genetic marker for increased risk of developing ovarian cancer, and they suggest that the KRAS-variant may be a new genetic marker of cancer risk for hereditary breast and ovarian cancer families without other known genetic abnormalities."

What we now need is validation in large scale clinical trials to determine whether the genetic marker can be used commercially to determine prognostic risk for ovarian cancer earlier.  If the results ultimately prove useful in clinical trials, then this finding relating to the KRAS variant may well have important implications for future therapeutic strategies and pipeline development in ovarian cancer as well as for prognostic testing and earlier detection for improved outcomes. 

ResearchBlogging.org
Ratner, E., Lu, L., Boeke, M., Barnett, R., Nallur, S., Chin, L., Pelletier, C., Blitzblau, R., Tassi, R., Paranjape, T., Hui, P., Godwin, A., Yu, H., Risch, H., Rutherford, T., Schwartz, P., Santin, A., Matloff, E., Zelterman, D., Slack, F., & Weidhaas, J. (2010). A KRAS-Variant in Ovarian Cancer Acts as a Genetic Marker of Cancer Risk Cancer Research DOI: 10.1158/0008-5472.CAN-10-0689 

Quaye, L., Song, H., Ramus, S., Gentry-Maharaj, A., Høgdall, E., DiCioccio, R., McGuire, V., Wu, A., Van Den Berg, D., Pike, M., Wozniak, E., Doherty, J., Rossing, M., Ness, R., Moysich, K., Høgdall, C., Blaakaer, J., Easton, D., Ponder, B., Jacobs, I., Menon, U., Whittemore, A., Krüger-Kjaer, S., Pearce, C., Pharoah, P., & Gayther, S. (2009). Tagging single-nucleotide polymorphisms in candidate oncogenes and susceptibility to ovarian cancer British Journal of Cancer, 100 (6), 993-1001 DOI: 10.1038/sj.bjc.6604947

Chin, L., Ratner, E., Leng, S., Zhai, R., Nallur, S., Babar, I., Muller, R., Straka, E., Su, L., Burki, E., Crowell, R., Patel, R., Kulkarni, T., Homer, R., Zelterman, D., Kidd, K., Zhu, Y., Christiani, D., Belinsky, S., Slack, F., & Weidhaas, J. (2008). A SNP in a let-7 microRNA Complementary Site in the KRAS 3' Untranslated Region Increases Non-Small Cell Lung Cancer Risk Cancer Research, 68 (20), 8535-8540 DOI: 10.1158/0008-5472.CAN-08-2129

"Aprepitant is the first commercially available drug of a new class of neurokinin-1–receptor antagonists for treating chemotherapy-induced nausea and vomiting. The dominant ligand for the neurokinin-1 receptor is substance P. An increase in the number of neurokinin-1 receptors on keratinocytes has been found in patients with chronic pruritus." 

 Source: NEJM

This was an interesting Letter to the Editor in the New England Journal of Medicine that I came across while looking for a different letter on JAK2 mutations and coronary ischaemia.

Essentially, the Italian authors described a brief case study where they treated two patients who were being treated with erlotinib (Roche) for non-small cell lung cancer (NSCLC) developed pruritis, ie the classic acneiform rash associated with EGFR therapy. They were non-responsive to standard systemic treatment and topical glucocorticoids.

Treatment with erlotinib was temporarily stopped while aprepitant (Merck) was given for three days, leading to "a prompt recovery" from the pruritis. Unfortunately, on resuming erlotinib with prednisone and anti-histamine prophylaxis, the pruritis returned, so the two drugs were tried concomitantly for 2 months. The severe pruritis was successfully contained (although the rash remained) and tumour responses are now being assessed.

What's interesting about this report is that normally one would not normally think of a therapy for nausea and vomiting as treatment for pruritis, but in this case, it appears to be mechanism related, ie NK1 receptors in keratinocytes found in pruritis.  The idea for the treatment came from previous correspondence to the same journal late last year regarding pruritis associated with Sézary syndrome and T-cell lymphomas.

The acneiform rash is psychologically distressing to many patients who develop it on EGFR therapy, thus prophylactic therapy that enables them to complete their anti-cancer treatment may well be a good thing, especially if they see a tumour response and, hopefully, an improvement in outcome.

The other day as I was clearing out my office of a hideous amount of paper as we continue our quest for a true minimalist and paperless office that is also scanned and searchable as well as being environment friendly, when I came across a tiny scrap of newspaper. Before throwing it out, curious, I wondered what it was about as a large starfish in the centre caught my attention.

It was a clipping from the Mail on Sunday (oops) back in early 2008.

Verona The snippet described how a starfish could potentially point to a new way of developing treatment for asthma. The company involved, Verona Pharma, was headed by Clive Page, a Pharmacology Professor from Kings College London, where I did my Ph.D research in asthma back in the late 80's (hello, Clive!). We met at a fascinating lecture at the Brompton Hospital given by Prof Peter Barnes, I recall. No wonder I tore out the article in the financial section after idly picking up the paper on the Tube, it's like finding old memories in sepia print.

Serendipity is a fine thing sometimes.

Wondering if the company was still going (they were looking for approval for a clinical trial at that time according to the newspaper), I checked them out and was delighted to see they're not only still going strong, but now have 3 products in development and are doing well, including RPL554, the compound mentioned in the clipping.

The late 80's and early 90's were almost a heyday for respiratory research. Many of the University of London respiratory labs (including Kings and the Brompton) were busy doing large scale clinical trials that later led to a new raft of inhibitors as either long-acting beta2-adrenergic agonists eg bronchodilators (salmeterol) or new generation steroidal anti-inflammatories (fluticasone) for treating asthma. There was also a vibrant basic research community at that time looking at phosphodiesterase (PDE) 3 and 4, leukotrienes (LT), prostaglandins, cytokines and thromboxanes as mediators of the inflammatory response.

It was therefore very interesting to me to discover that Verona's lead compound, RPL554, is a mixed phosphodiesterase (PDE) 3 and 4 inhibitor (a bronchodilator and anti-inflammatory compound in a single molecule). Furthermore, it was co-invented by Sir David Jack (former Research Director of Glaxo) who was involved in the development of the inhaled asthma drugs such as salmeterol and fluticasone, among others.

Meanwhile, I was pleased to see on Verona's website that RPL554 has completed a phase I/IIa trial of RPL554, and will watch it's future progress with interest. The company is taking a smart approach to drug development. Rather than putting all their eggs in one pipeline basket,Verona have have several compounds in preclinical development for related areas such as cough and have also identified some marine organisms (hence the starfish in the news clipping), a natural source of novel anti-inflammatory polysaccharide (NAIPS) without unwanted anti-coagulant actions.

It's going to be interesting to watch Verona's development and I hope at least one of the compounds makes it to market because the underlying scientific rationale is solid.

Watch this space!

We enjoy receiving your comments on the blog or the many that come by email.  We respect your privacy and do not add people to lists.  

A number of people have asked how to subscribe to Pharma Strategy.  If you enjoy reading this blog, you can add it to your RSS reader using the orange logo, bookmark it or sign up below and receive updates to your inbox:

Enter your email address:

Delivered by FeedBurner

1 Comment

Today I will be out of the office at client meetings, but many of you will be wondering what will happen at the FDA ODAC briefing meeting, which will be discussing the bevacizumab (Avastin) data in breast cancer this morning. You can take a look at the briefing documents here.

Background

Avastin was approved to much fanfare because it was basically shown to shrink tumours quite dramatically and the early magnitude of the PFS data suggested benefit in favour of adding Avastin to chemotherapy.  According to the FDA, full approval would be thus debated once the final survival data were available:

“As a condition of the accelerated approval, Genentech was required to submit data from two ongoing, placebo-controlled trials (AVADO and RIBBON1) to provide verification of the treatment effect on PFS and to provide additional information on the effects on overall survival.”

According to the FDA documents:

“The addition of bevacizumab to paclitaxel resulted in a 52% increase in progression-free survival (HR 0.48, 95% CI 0.39, 0.61; p< 0.0001) with an observed 5.5-month difference in median PFS, based on an independent radiographic review.

There was no significant difference in overall survival, a secondary endpoint, between the two treatment arms. The tumor response rate was higher with bevacizumab plus paclitaxel as compared to paclitaxel alone (48.9% versus 22.2%).”

Breast cancer was one of the early tumour types that Genentech tested the drug in but despite promising phase II data, the phase III study was negative and thus they decided to go back to the drawing board with the statisticians and investigate the drugs safety and efficacy in another large scale phase III trial.  Meanwhile, colon cancer overtook breast cancer and became the first approved indication for Avastin instead.

Fast forward to 2010.

The data from AVADO and RIBBON1 is now available in the FDA briefing documents. Here is a quick synopsis from each.  I confess that I found the FDA summary of the data a little confusing, but the tables tell the picture more easily and have included the efficacy data tables from the briefing documents, where appropriate.

AVADO

This study comprised a double-blind, placebo-controlled, three-arm trial of:

  • docetaxel plus placebo
  • docetaxel plus bevacizumab 7.5mg/kg
  • docetaxel plus bevacizumab 15 mg/kg

with all therapies given on a standard three-weekly schedule.

A total of 736 patients with HER-2 neu negative tumors who had not received prior chemotherapy for metastatic breast cancer were enrolled.

Overall the data was summarised as follows:

Picture 3
Source: FDA (pdf link)

The OS Kaplan-Meier curve for the AVADO study looks like this – you can see that they cross over just coming up to two years:

image from img.skitch.comSource: FDA (pdf link)

The FDA went on to describe the tolerability data as thus:

“Safety data showed an increase of grade 3-5 adverse events, serious adverse events and study drug discontinuation with the addition of bevacizumab to docetaxel.  More patients in the bevacizumab-containing arms required interruption/dose reduction or discontinuation of docetaxel due to an adverse event.”

RIBBON1

This was a double-blind, randomized, parallel group study conducted in people with metastatic or locally recurrent HER2-neu negative adenocarcinoma of the breast, who had not received prior chemotherapy for their advanced or metastatic cancer.  A total of 1237 patients were randomized (2:1) to receive anthracycline or taxane-based chemotherapy (n=622) or capecitabine (n=615) in combination with either bevacizumab or placebo.

The efficacy data looks like this:

Picture 4Source: FDA (pdf link)

The FDA also summarised the side effect profile:

“Overall, the incidence of grade 3-5 AEs and serious AEs were almost twice as high in the bevacizumab arms compared to placebo arms in both cohorts. In the taxane/anthracycline cohort, taxane subgroup, there was slightly more deaths in the bevacizumab containing arm than placebo arm (49.8 % versus 43.1 %).

The vast majority of the deaths were attributed to breast cancer. Adverse events known to be caused by bevacizumab were, as expected, increased in the bevacizumab containing arms in both cohorts. The most common AEs associated with bevacizumab were hypertension, bleeding/hemorrhage and febrile neutropenia.”

Essentially though, the incidence of AEs is not significantly different than currently described in the prescribing information.

Overall Conclusions:

In the summing up to the ODAC committee, the FDA briefing document declares:

“AVADO and RIBBON 1 are well conducted, double-blinded trials.  The magnitude of the improvement in PFS observed in these two studies failed to confirm the magnitude of PFS improvement observed in the E2100 trial, the basis for the accelerated approval.

The magnitude of treatment effect is clinically important because it is really a measure of delaying symptoms from tumor progression, which has to be weighed against drug toxicity that is present for the duration of treatment.”

The short answer is that the overall survival data suggested hazard ratios favoring the placebo arms in the AVADO study and the taxane/anthracycline cohort of the RIBBON1 study.  This is important, because it raises the issue of risk:benefit because the women are being exposed to significant toxic side effects with no impact on overall survival.

What next?

Looking at the data from the two trials objectively, it is hard to justify treating women with newly diagnosed breast cancer with Avastin, when they would appear to live longer and have less side effects with chemotherapy alone.  The PFS effect seen in E2100 has not been confirmed at all.

Ultimately, I think Roche/Genentech would have a much better case to argue to the FDA and public if they had a biomarker to determine which patients are likely to do better on bevacizumab therapy, just as erlotinib works better in lung cancer patients who are EGFR mutation positive.

I wonder what ODAC will decide?

My educated guess is that one of two scenarios might happen:

  1. Approval remains, but modify the PI language similarly to Iressa to indicate that there is no overall survival advantage associated with adding Avastin to chemotherapy in breast cancer.
  2. Withdrawal of breast cancer indication for Avastin.

Technically, logic would suggest the latter option is more likely given the rules governing accelerated approval, but these panels are a notorious minefield sometimes, very hard to predict and the FDA doesn’t always follow their advice, as the original approval for Avastin in breast cancer demonstrated!

What do you think?

{UPDATE: My educated guess was correct – ODAC voted 12-1 to withdraw Avastin from the market.  The big question now is what will the FDA decide?}

{UPDATE 2: FDA decided to hold a public hearing, now scheduled for Weds June 29th, 2011}

3 Comments

Recently at a couple of scientific cancer meetings, American Urology Association (AUA) and American Society of Clinical Oncology (ASCO), Frank McCormick described a fascinating talk about how a wac-a-mole approach to figuring out how the phosphatidylinositol 3′-kinase (PI3-kinase or PI3K) pathway could be targeted effectively with therapeutics. The reason for research in this area is PI3K has been shown to play a major role in proliferation and survival in a wide variety of human cancers, thus making is a potential target for therapeutic intervention.

I’ve been following this target for a couple of years now and data is now starting to emerge that’s worth discussing on a broader scale, given the implications.  Here’s a quick snapshot of the PI3K pathway and related pathways:

image from www.nature.com
Source: Workman et al., Nature

As many of us well know, however, simply targeting one element of an aberrant pathway can lead to cross-talk and feedback loops as the cancer tries to maintain the signals important for it’s survival, so a more cunning approach is needed whereby the escape routes are closed off one by one by targeting different kinases as well as PI3K.

McCormick’s talk was a fascinating lecture that basically went through multiple pathways explaining, ‘well we tried X and this happened, so we tried blocking Y as well and this happened…’  kind of approach in a very logical and systematic fashion.  Eventually, all options will be explored and a new paradigm might emerge.

It was therefore with great interest that I read a series of new papers in AACR’s journal, Clinical Cancer Research (see references below) over the weekend on both the pathway itself, and also new data with targeted PI3K agents in both breast and renal cancers.

The Data so far:

Miron et al., looked at PI3K mutations in in situ and invasive breast carcinomas and reported:

“This is the first study to show that PIK3CA mutation is a relatively early event in breast tumorigenesis preceding invasion because the frequency of PIK3CA mutations was the same in pure DCIS as in DCIS adjacent to IDC and in IDC.”

Given the frequency of mutations was the same for the 3 groups they studied (pure ductal carcinoma in situ (DCIS), DCIS adjacent to invasive carcinoma, and invasive ductal breast carcinomas), the data suggest that the PI3K mutation may play a greater role in breast tumor initiation than in invasive progression.

If this is the case, targeting PI3K early, for example in neoadjuvant therapy, may have a positive beneficial effect.

In the O’Brien paper, the researchers looked for predictive biomarkers of sensitivity to Roche/Genentech’s PI3Ki, GDC-0941 in preclinical models of breast cancer:

“We found that models harboring mutations in PIK3CA, amplification of human epidermal growth factor receptor 2, or dual alterations in two pathway components were exquisitely sensitive to the antitumor effects of GDC-0941.  We found that several models that do not harbor these alterations also showed sensitivity, suggesting a need for additional diagnostic markers.”

Identifying suitable biomarkers in preclinical studies, such as the HER2 amplification and the PIK3CA mutation (but not PTEN deficiency) previously identified in other studies and now validated in O’Brien et al’s GDC-0941 study, will hopefully help in better design of future clinical studies.  They also noted that decreased ERBB3 expression in PIK3CA mutant cell lines, and ERBB3 expression was increased in response to treatment with a PI3K inhibitor, suggesting that ERBB3 expression levels might be used as a biomarker for high activation of PI3K signaling and increased sensitivity to PI3K inhibitors.  This kind of rigourous approach would potentially enable selecting which people are most likely to respond up front to the agent, rather than exposing those who are unlikely to get a response to additional toxicities and side effects.

In a well written editorial, Turke and Engelman, also emphasised that:

“A novel expression profile was developed to identify other breast cancers sensitive to PI3K inhibitors. These expression studies highlighted feedback networks connecting TORC1, PI3K, and mitogen-activated protein kinase (MAPK) pathways, and underscored the potential for combination therapies.”

They also went on to observe:

“It will be interesting to determine if PI3K inhibitors induce substantial apoptosis in vitro and tumor regressions in vivo in these cancer models (without HER2 amplification or PIK3CA mutation).  Of course, it will be crucial to assess biomarkers identified in laboratory studies in clinical samples from patients who respond to PI3K inhibitors.  Neo-adjuvant trials in breast cancer patients can be leveraged to address these translational goals, because they correlate clinical efficacy and pathologic signs of response (e.g., changes in Ki67 levels and induction of caspase cleavage) with the presence of potential biomarkers.”

In another study, Cho et al., looked at the effects of a dual PI3-Kinase/mTOR Inhibitor
NVP-BEZ235 compared with rapamycin in renal cancer (RCC) with BEZ235 (Novartis). The proof of concept for mTOR has already been shown clinically with the approval of two drugs in this indication, temsirolimus (Pfizer) and everolimus (Novartis):

“These agents induce only modest tumor regression and extend progression-free survival only a few months in most patients.”

The big question here is whether targeting PI3K as well as mTOR would have any extra beneficial effects?  The results demonstrated that dual inhibition of PI3K/mTOR with BEZ235 induced growth arrest in RCC cell lines both in vitro and in vivo more effectively than inhibition of TORC1 alone. If reproduced in the clinic, this may offer a new and more effective approach to treatment of the disease.

The Future:

The PI3-kinase field is particularly interesting, with several companies snapping up PI3K inhibitors including sanofi-aventis (from Exelixis) and more recently, Infinity (from Intellikine).  Other oncology companies already have some in their pipeline, such as Novartis (BEZ235) and Roche/Genentech (from Piramed).  Meanwhile, smaller biotechs such as Semafore and Calistoga also have some promising early phase compounds in development.  Some of these compounds target PI3-kinase alone, while others target PI3K and mTOR.

This is not going to be a straightforward approach to targeting cancer and identifying biomarkers along the way will be key, as well working out the best combinations that might make a more effective therapeutic approach than single agent activity. Figuring out when best to test these agents (early or late) will also be critical. The I-SPY breast cancer trials have already led the way in creating protocols for testing novel agents in the neoadjuvant setting in breast cancer, and it may well be that PI3K inhibitors would be a good class to test in this setting based on the new evidence from Miron et al’s study.

What is particularly interesting to me is that PI3K signalling may also have a role to play in asthma and COPD (the area I did my doctoral research in) rather than just cancer.  Now that would be really fascinating as the biochemical and molecular biology overlap have long been suspected, but very little research has really evolved this way. Part of that is due to drug manufacturer silos and the inability to effectively spearhead cross-therapeutic research.

It will be fascinating to watch how the PI3K data shakes out in practice over the next few years.

What do you think?

ResearchBlogging.org O’Brien, C., Wallin, J., Sampath, D., GuhaThakurta, D., Savage, H., Punnoose, E., Guan, J., Berry, L., Prior, W., Amler, L., Belvin, M., Friedman, L., & Lackner, M. (2010). Predictive Biomarkers of Sensitivity to the Phosphatidylinositol 3′ Kinase Inhibitor GDC-0941 in Breast Cancer Preclinical Models Clinical Cancer Research, 16 (14), 3670-3683 DOI: 10.1158/1078-0432.CCR-09-2828

Turke, A., & Engelman, J. (2010). PIKing the Right Patient Clinical Cancer Research, 16 (14), 3523-3525 DOI: 10.1158/1078-0432.CCR-10-1201

Miron, A., Varadi, M., Carrasco, D., Li, H., Luongo, L., Kim, H., Park, S., Cho, E., Lewis, G., Kehoe, S., Iglehart, J., Dillon, D., Allred, D., Macconaill, L., Gelman, R., & Polyak, K. (2010). PIK3CA Mutations in In situ and Invasive Breast Carcinomas Cancer Research, 70 (14), 5674-5678 DOI: 10.1158/0008-5472.CAN-08-2660

Cho, D., Cohen, M., Panka, D., Collins, M., Ghebremichael, M., Atkins, M., Signoretti, S., & Mier, J. (2010). The Efficacy of the Novel Dual PI3-Kinase/mTOR Inhibitor NVP-BEZ235 Compared with Rapamycin in Renal Cell Carcinoma Clinical Cancer Research, 16 (14), 3628-3638 DOI: 10.1158/1078-0432.CCR-09-3022

9 Comments

Twitter is great for highlighting interesting journal articles, as I found when Edward Winstead from the NIH shared this paper from PLOSone on the importance of microRNA in melanoma in his Twitter stream (thanks, Ted!).

image from en.wikipedia.orgThere has been a lot of interest in melanoma lately, with the rise of a couple of interesting new compounds targeting different mutations or kinases including CTLA4 by ipilimumab (BMS) and B-RAF by PLX-4032 (Plexxikon/Roche).  

You can see some of the recent data I've blogged about herehere and here.  

At the moment, we're waiting for the new data from PLX-4032 at a melanoma conference later this year and BMS may be filing their phase III data in ipilimumab by the end of this year after some promising reactions to the data presented last month in the plenary session at ASCO.  In addition, GSK also have some compounds in earlier development that are generating interest.

How does the new data in PLOSone connect with melanoma?

Well, it's an aggressive and highly malignant cancer and scientists have long wondered how melanoma cells travel from primary tumours on the surface of the skin to the brain, liver and lungs, where they become more aggressive, resistant to therapy, and deadly. This ultimately makes treatment and control of the disease very challenging.

In Feb 2009, an article in PNAS (see reference below) suggested that the culprit might be a short strand of RNA called microRNA (miRNA) that is over-expressed in metastatic melanoma cell lines and tissues. It is also known that the Microphthalmia associated transcription factor (Mitf) is an important regulator in melanocyte development and has been shown to be involved in melanoma progression.
The new data reported in PLOSone this month takes our understanding a little further and shows that microRNAs are also involved in regulating Mitf in melanoma cells. 

The authors concluded that:

"miR-148 and miR-137 present an additional level of regulating Mitf expression in melanocytes and melanoma cells. Loss of this regulation, either by mutations or by shortening of the 3′UTR sequence, is therefore a likely factor in melanoma formation and/or progression."

What this means is that the microRNA's involved may offer new therapeutic targets in order to either reduce the development of resistance or aggressive progression and metastasis (ie spread of melanoma) to other organs. In order words, future research may involve the addition of microRNA therapy to optimise outcomes.

For now, microRNA is very much a research on the rise but it won't be long before we start seeing the first RNA based therapies in the clinic based on a solid scientific research rationale.  As our understanding of the complex biology improves, so does the chances of developing a multi-factorial strategy to combat the devastating disease.

For those of you interested in this exciting field, I'll cover a more basic primer on microRNA and RNA therapeutics in development in a future blog post.

Photo Credit: Wikipedia

ResearchBlogging.org
Haflidadóttir, B., Bergsteinsdóttir, K., Praetorius, C., & Steingrímsson, E. (2010). miR-148 Regulates Mitf in Melanoma Cells PLoS ONE, 5 (7) DOI: 10.1371/journal.pone.0011574

Segura, M., Hanniford, D., Menendez, S., Reavie, L., Zou, X., Alvarez-Diaz, S., Zakrzewski, J., Blochin, E., Rose, A., Bogunovic, D., Polsky, D., Wei, J., Lee, P., Belitskaya-Levy, I., Bhardwaj, N., Osman, I., & Hernando, E. (2009). Aberrant miR-182 expression promotes melanoma metastasis by repressing FOXO3 and microphthalmia-associated transcription factor Proceedings of the National Academy of Sciences, 106 (6), 1814-1819 DOI: 10.1073/pnas.0808263106

error: Content is protected !!