Biotech Strategy Blog

Commentary on Science, Innovation & New Products with a focus on Oncology, Hematology & Cancer Immunotherapy

Posts from the ‘Translational Medicine’ category

2012 Annual meeting of American Association for Cancer Research in Chicago. Photo Credit: Pieter DroppertAs Sally Church, PhD noted on Pharma Strategy Blog, the 2012 annual meeting of the American Association for Cancer Research (AACR), recently held in Chicago, showcased many new cancer products in early development.

Cancer new products have a high attrition rate as they move through the development pipeline, so any promising results seen in early stages of development must be viewed with caution.

Results from laboratory studies using cell lines or trials in animals do not always translate into new drugs that work in man, e.g. they may have an unacceptable toxicity, not target the driver mutation, or adaptive resistance may just lead to the cancer bypassing the blocked pathway.

However, scientific meetings such as AACR do provide a window into the possible new drugs of the future. One prostate cancer new product that caught my attention at AACR 2012 as one to watch is AZD3514.

Sarah Loddick from AstraZeneca gave one of the few oral presentations at AACR on this exciting new compound.  This was the only AACR session I attended where I was able to access wifi. Some of my live-tweets are captured in the Storify below (click here to access this on Storify):

http://storify.com/3nt/aacr-2012-azd3514-in-prostate-cancer

Unfortunately, Sarah Loddick has not (as of time of writing) shared a copy of the AZD3514 prostate cancer poster that she presented later in the meeting, so I’m unable to write more about the preclinical prostate cancer data.

AZD3514 is a novel selective androgen receptor down-regulator (SARD) and has a different mechanism of action to drugs such as enzalutamide (MDV3100) that functionally inhibit AR signaling by binding to the AR & AR splice variants.

Sarah Loddick concluded at the end of her oral presentation that AZD3514:

  • inhibits prostate cancer growth in vitro & in vivo
  • has activity against wild-type and mutated AR
  • has activity in pre-clinical models that represent castration resistant prostate cancer (CRPC)
  • inhibits seminal vesicle growth in rats in the presence of physiological levels of circulating tumor cells.

AZD3514 is in a multi-center phase 1 clinical trial in patients with metastatic CRPC in Europe (NCT01162395) and Japan (NCT01351688). I look forward to seeing the presentation of the results from these trials.

From what I saw at AACR, AZD3514 is a new prostate cancer drug to watch.

Update April 20, 2012

I was delighted to receive an email this morning from Sarah Loddick of AstraZeneca with a copy of the AZD3514 poster that I requested (AACR abstract #3848): “Pre-clinical profile of AZD3514: a small molecule targeting androgen receptor function with a novel mechanism of action and the potential to treat castration resistant prostate cancer.

I am sensitive to the unpublished status of much of the research presented at AACR, but without giving too much away, some of the key messages from this poster are that AZD3514:

  • Binds to the androgen receptor (AR) ligand binding domain & reduces viability of prostate cancer cells in vitro. 
  • Inhibits AR transcriptional activity within 2h of exposure in LNCaP cells, and reduced both PSA & TMPRSS2 mRNA
  • Inhibits AR induced translocation to the nucleus
  • Causes AR down-regulation in prostate cells in vitro
  • Causes AR down-regulation in rat R3327H prostate tumors
  • Has activity in pre-clinical models of CRPC

A drug such as AZD3514 in prostate cancer could potentially be used to overcome resistance to enzalutamide (MDV3100), or alternatively it could be used ahead of enzalutamide if it has the potential to avoid resistance and offer better outcomes. We obviously will have to wait for clinical data to see what it’s true potential is and the data from AACR, while promising, is still only preclinical.

The prostate cancer market is a busy one and companies with AR targeted new products in development will have to offer drugs that are superior to enzalutamide if they wish to have lasting commercial success.

Update June 6, 2013: AstraZeneca terminates development of AZD3514 in Advanced Prostate Cancer

At ASCO 2013 it was announced that the development of AZD3514 in advanced prostate cancer has been terminated. You can read more about what happened in the first-in-human clinical trial in my AZD3514 blog post from ASCO 2013.

1 Comment

Maha Hussain MB ChB is Professor of Medical Oncology at the University of Michigan. I was privileged to interview her about cabozantinib (XL184), a novel drug in development for multiple indications, including prostate cancer.

You can read part 1 of the interview on cabozantinib and pain here, and part 2 about the bone effect seen with cabozantinib, here.

In this final post, Dr Hussain discusses the scientific rationale behind the mechanism of action for cabozantinib, a multi-kinase inhibitor of C-MET, VEGF and others.  She also shares her thoughts on where the scientific data suggests it should have most impact on prostate cancer.  I have emphasized in bold a few sections that stood out to me.

BSB:  We saw that the VEGF inhibitor bevacizumab (Avastin) failed in prostate cancer, so what is the scientific rationale behind targeting c-MET & VEGF? 

Dr Hussain: Let me just begin with a little background. The c-MET pathway and VEGF pathway from a generic perspective in cancer are specifically of interest.  The trial [with cabozantinib] was originally designed, not that there was any specific preclinical data with the drug in prostate cancer, but rather because of the fact that the MET signalling pathway appears to be relevant in a variety of tumors where there is an overexpression.  This potentially causes us, or at least the sponsor I should say, to believe this is worth targeting.

In retrospect, the sponsor targeted diseases where there seems to be overexpression of MET and that’s how it started.  Now, there is data though in terms of overexpression, if you looked at tissues from primary versus lymph node versus metastatic disease there appears to be a stepwise increase in the rate of overexpression of MET in specifically prostate cancer.  

In the short audio clip below Dr Hussain next discusses the key finding that if you block Androgen Receptor (AR) signaling, MET expression goes up. Click here if you can’t see the SoundCloud audio player.


Dr Hussain continuesWhat happened is serendipity occurred, and that is the drug appeared to be very active.  In essence, we are now going back to the lab to try to figure out exactly why does it work?  Is it just a bone only issue, and when I say bone I mean microenvironment issue, or is it actually more than that, is it a totality of both microenvironment and an anti-tumor effect?

What’s interesting is in the literature, there is data that would indicate that when you activate MET, one protects the cancer cells from potential damaging effects of DNA damaging agents. There is also some data, not in prostate cancer specifically, that MET actually mediates resistance to other growth factors like EGFR and Src and so on.  When you try to inhibit them, MET actually appears to potentially mediate resistance to those inhibitors.  There is also data that potentially MET pathway stimulates angiogenesis in the context of sunitinib resistant tumors.

Now in prostate cancer, what we we know is that VEGF is critical for the osteoblast, osteoclast action, and both MET and VEGF seem to crosstalk to regulate the interaction between the tumor cells, the bone cells, the endothelial cells and so on. There are several trials that have targeted the microenvironment. 

You mentioned Avastin, but the reality of it is that sunitinib was a negative trial also, and  the bone targeted drugs have been generally negative trials. 

The question is, why does this thing work when everything else has failed?  My guess is that it may very well may be that pure targeting of one pathway is not enough and that you need a multi-targeted approach. 

The drug, while it is targeting VEGF and MET, it is not a pure target of VEGF and MET, it has multiple other effects, and truthfully no one can say the reason it is working in prostate cancer is because it is inhibiting MET.  I honestly don’t think that we have demonstrated that. 

We do have a clinical trial actually that has just started to specifically look at what happens in the tumors in the bone in patients who are going to be treated with this drug.

BSB:  Did you see the recent paper in Cancer Discovery that talked about suppression of tumor invasion and metastasis by concurrent inhibition of C-MET and VEGF signaling in pancreatic neuroendocrine tumors? 

[As an aside for those interested, Sally Church, PhD has an excellent post on Pharma Strategy Blog, Combined VEGF and MET inhibition in some cancers may be better than either alone,” which discusses the Cancer Discovery paper by Sennino et al.)

Dr Hussain:  In my [AACR] presentation, I do put the data from the pancreas neuroendocrine [research]. It is not from that paper, but I think it is data from previous papers that were published that looked at how the drug works. This was data that I was able to get through the sponsor from Drs Sennino and McDonald.

BSB: Would we we better off combining separate c-MET and VEGF inhibitors rather than have one drug like cabozantinib that has multiple targets?

Dr Hussain: I think you want to go with what works and thus far, this is the first drug that has again what we think is a MET and VEGF targeting based on the preclinical data, that seems to work.  But I would also point that it is not a pure VEGF & MET it also has RET, KIT, FLT and AXL.  My point here is, we assume that this is the reason.

But I would point out is, that I think the notion that you are going to declare victory on cancer by targeting one pathway in a disease that has multiple redundant pathways, in my view is a naive assumption.  We have known for example, with even diseases that are chemosensitive that you couldn’t cure them with one drug and Testes cancer is a perfect example.  Probably, the model of a curable cancer is Testes cancer. Because there is no other cancer, a solid tumor that you actually target, treat in metastatic disease and can cure at a high rate.  There is just nothing else, with all these new discoveries with targeted drugs, none of them have thus far cured a metastatic patient.  And I would stand corrected if I am not right.

BSB: So your suggestion is that we will need to combine these drugs?

Dr Hussain:  Absolutely! The problem is if you are a scientist and you put in for a grant and you put in a drug that is quote unquote a “dirty drug” because it is has multi-targeting, that is not scientifically good enough. Yet from a clinical perspective, I think that what is clear is we are not curing people by having one target targeted. I would argue that all of the “progress” that has occurred if you look at the drugs, they are targeting pathways that are isolated pathways that are not necessarily highly prevalent in all diseases. In some diseases it is a small percentage of patients that have those pathways.

If you look at the outcomes, these are clearly not what I would call huge victory, because you are basically either causing a response or a short lived remission and not significantly impacting survival, as in curing cancer, prolonging life by years.  So I go back to the model of Testes cancer, which is again one of the most curable cancers that we have in the metastatic disease setting.  And even then, we do not cure it with one drug.

BSB:  Can I take you back briefly to something you said earlier that caught my attention. You said earlier that expression of c-met was increased after attenuation of the androgen receptor signaling, so one strategy would be to inhibit activation of both the c-MET and AR signaling pathways.

Dr Hussain:  Correct, correct.

BSB:  I saw that there is a phase 1 trial just starting looking at abiraterone in combination with cabozantinib, so is the theory there that we could potentially delay time to castration resistance if we inhibit both AR signaling and the c-met/VEGF pathway?

Dr Hussain:  Correct, but I would say that the place to start is not with abiraterone, but with primary hormonal treatment when the tumor is still sensitive to hormones.

BSB: So when we are doing androgen deprivation therapy?

Dr Hussain: Correct. I personally think that is where we need to start.  And in fact because of that, in partnership with one of my colleagues here who is also my co-leader for the prostate cancer program at Michigan, we have some preclinical data that is in progress looking at hormone sensitive tumor model.

The question comes up where is the right time to put these drugs, and the truth of it is you brought up the issue of expense, and while I am not the person who says how much life is worth, clearly if it is my life or my family’s life or my patients life, it is worth a lot. But society has to decide how much money to spend on what and for what magnitude of benefit. That is something that I think we are all struggling with, as you point out, because healthcare dollars are going down and a lot of these drugs are very expensive. And when they work we talking about prolongation of life anywhere from 2 months to 5 months, and that’s average. The reality of what that means is that you are treating thousands of patients to get benefits in a smaller number of patients, and is this something sustainable and is this the way we should go?

I would argue that, life is very precious and very valuable, the more that we can do to impact it, the better we will be as doctors, patients and society.  But that in order to get a bigger bang for the bucks, so to speak, for the drug itself is we need to begin to think about moving these drugs in settings whereby you have cancers that are not as resistant where the impact might be higher.  Imagine then, we would then prolong life by maybe years, as opposed to months.  I think that would be much more worth it.

Yes, it will be more costly to test these drugs earlier, and certainly there is the financial aspect of it for Pharma. They would want to go in a setting where they can get the results sooner rather than later, and not treat ten times more patients to find the results. I would say from the big picture perspective as a doctor, that’s not my focus, my focus is to move these drugs to settings where we can have more impact for more patients, and more meaningful impact on their outcomes.

BSB: In essence, you do think that is where cabozantinib could have most effect at that time?

Dr Hussain:  Maybe. I think this is a question that should be asked. We have proposed to look at in the preclinical setting, to try to justify a clinical trial potentially.  Recognizing that it is one of those things, clearly we are limited by access to drugs and $ to support these type of questions. And if it is not cabozantinib, it’s the next in line targeting that pathway. I do think someone needs to ask that question and we could envision trial designs that can give you short-term answers that will give you a sort of Go/No-Go type strategy.  It is a complex process, obviously. I would say if the world was perfect, and if it was my money and my drug, I would go that way to see how it behaves.

BSB: Thank you!

This brings to an end the series of posts covering my interview with Dr Maha Hussain, a thought leader in prostate cancer clinical research. I would like to sincerely thank her for the time she took out of her busy schedule to talk to Biotech Strategy Blog.

To effectively treat prostate cancer will require drugs to be given in combination. A VEGF/MET inhibitor may have a role to play in the treatment of prostate cancer, but whether Exelixis have the appropriate clinical trial strategy in place with cabozantinib to achieve this, remains to be seen.

Other pathways that we didn’t have time to discuss may also need to be targeted e.g. Src and PI3-Kinase.  It will be interesting to see what new data is presented at the annual meeting of the American Association for Cancer Research (AACR) in Chicago later this month.

Contrary to popular opinion, innovation is not dead in the biomedical industry, as evidenced by news of a novel drug-delivery system published as a Rapid Publication in Science Translational Medicine (STM) on February 16, 2012.

The paper from Robert Farra of MicroCHIPS, Inc. and research collaborators, describes a first-in-human testing of a wirelessly controlled drug delivery microchip.

Farra et al., report the results of a clinical trial with 8 women in whom microchips were implanted for 103 days. The data showed that the pharmacokinetic profile of microgram-quantities of the anti-osteoporosis drug, teriparatide (FORSTEO), delivered by the microchip was similar to subcutaneous injections.  However, the device did fail in one of the 8 women, so data is only reported for 7 patients, a very small patient sample.

Picture Credit: MicroCHIPS, Inc.

The drug delivery device is an array of 600-nL micro reservoirs in which the drug is stored, that is associated with a 13.0 mm x 5.4mm x 0.5mm silicon chip.

The microchip was implanted beneath the skin (subcutaneously) in the abdomen by creating a 2.5cm incision, performed during an outpatient visit.

This paper is also interesting for its use of telemedicine. A remote operator was able to establish a wireless link and send instructions directly to the implant on dosing schedule as well as receive information back on operation of the chip.

John T. Watson, Professor of Bioengineering at the University of California San Diego  commented in the accompanying editorial that:

“The microchip represents more than 10 years of engineering design and development efforts to arrive at a programmable, implantable device for subcutaneous release of a therapeutic agent in discrete doses.”

Multiple engineering design advances were made along the way.

He also noted the results from the quality-of-life surveys administered during the trial; the majority of women stating they often forgot they had the device implanted and would readily consent to a fresh implant if needed.

Innovations in drug delivery offer hope of an improved quality of life to patients with chronic disease who require daily injections.  In 2010, there were approximately 50,000 teriparatide users, not an insignificant market opportunity.  People with diabetes who require daily injection of insulin is another potential market that springs to mind.

The first-in-human results reported in Science Translational Medicine show promise and the potential of a novel implanted wireless drug delivery system.

However, many questions remain unanswered by this research including the reliability & durability of the microchip device, given that it failed in 1 out of 8 women implanted.

Further work on validating the technology, and confirming its safety, reliability and efficacy in a larger sample size will be needed before it can obtain regulatory approval.

References

ResearchBlogging.orgFarra, R., Sheppard, N., McCabe, L., Neer, R., Anderson, J., Santini, J., Cima, M., & Langer, R. (2012). First-in-Human Testing of a Wirelessly Controlled Drug Delivery Microchip Science Translational Medicine DOI: 10.1126/scitranslmed.3003276

Watson, J. (2012). Re-Engineering Device Translation Timelines Science Translational Medicine DOI: 10.1126/scitranslmed.3003687

One of the challenges of the next decade in cancer research will be targeting cancer metabolism; imaging is likely to play a key role in drug development.

NMR-image-of-brain-gliomaThe cover of the January 11 online issue of Science Translational Medicine (STM) shows a brain tumor (glioma) in red, detected using non-invasive nuclear magnetic resonance imaging that highlights cancer metabolism.

In a paper published in STM, Andronesi and colleagues from Harvard & other Cambridge, MA institutions (including Agios Pharmaceuticals – more on them later), showed that excess production of the metabolite 2-hydroxyglutarate (2HG) could be used as a biomarker for a subset of glioma.

The subset this metabolic biomarker identified, were those patients with mutations of the isocitrate dehyrogenase gene (IDH1), present in 86% of the grade II & III gliomas and secondary glioblastomas.

Agios Pharmaceuticals founded by eminent cancer researchers, Lewis Cantley, Tak Mak and Craig Thompson is targeting the IDH1 and IDH2 metabolic pathways.

They have shown that mutations of the metabolic gene IDH1 are consistent with that of a cancer-causing oncogene.  Interestingly, Agios notes on their website that IDH1 and IDH2 mutations have also been seen in acute myeloid leukemia (AML).

What makes 2HG a functional biomarker for glioma is its correlation with survival.  2HG accumulates in the brains of patients with IDH1 mutations. These patients have a greater survival than those with wild-type IDH1 gliomas.

Developing a drug that targets cancer metabolism in the brain is not easy. NMR imaging of the 2HG in the brain will help researchers non-invasively follow the effects of inhibitors of mutated IDH1. This is particularly important given that, according to Andronesi et al,  “no report exists about increased D-2HG in the blood, cerebrospinal fluid, or urine of glioma patients with IDH1 mutations.”

The January 11 online issue of STM, also contains another paper on the detection of 2HG using NMR. Elkhaled and colleagues from UCSF report a technique of proton high-resolution magic angle spinning spectroscopy.  Their data confirms the potential of 2HG as a surrogate marker of patient survival.

Cancer metabolism as a drug development target is an area I expect we will see more of in the next ten years.  Key to success will be the ability to identify biomarkers with which to assess and monitor the success of drug candidates.

The identification of 2HG as a biomarker for IDH1 in glioma patients shows that cancer metabolism is an area of potential for drug development.

One cloud on the horizon for Agios Pharmaceuticals is, however, the filing of a lawsuit late last year by the Abramson Cancer Institute of the University of Pennsylvania. This alleges that Craig Thompson concealed the start-up of Agios while working for Penn, and in essence took the intellectual property of the University to the company. The merits of this claim have yet to be decided.

References

ResearchBlogging.orgAndronesi, O., Kim, G., Gerstner, E., Batchelor, T., Tzika, A., Fantin, V., Vander Heiden, M., & Sorensen, A. (2012). Detection of 2-Hydroxyglutarate in IDH-Mutated Glioma Patients by In Vivo Spectral-Editing and 2D Correlation Magnetic Resonance Spectroscopy Science Translational Medicine, 4 (116), 116-116 DOI: 10.1126/scitranslmed.3002693

Elkhaled, A., Jalbert, L., Phillips, J., Yoshihara, H., Parvataneni, R., Srinivasan, R., Bourne, G., Berger, M., Chang, S., Cha, S., & Nelson, S. (2012). Magnetic Resonance of 2-Hydroxyglutarate in IDH1-Mutated Low-Grade Gliomas Science Translational Medicine, 4 (116), 116-116 DOI: 10.1126/scitranslmed.3002796

1 Comment

I have had the privilege to attend many scientific and medical congresses this past year, and my belief from listening to many presentations is that drug development innovation comes from understanding basic biology, then applying this knowledge.

Lisa Coussens (UCSF) at the 2011 San Antonio Breast Cancer Symposium (SABCS) provided a good example of how scientific knowledge is being translated into medicine and applied to drug development.

In her plenary presentation, she outlined how our understanding of the biology of macrophages and the importance they play in breast cancer may lead to new drug targets.  As an example of this, she showed pre-clinical animal work on the Plexxikon drug PLX3397.  A human phase 1b/II clinical trial will start in the near future.

I have aggregated some of the live tweets from the session using Storify.  Social media can be a powerful tool to share highlights and top-line messages with those not at meetings, as well as have a real-time conversation with those in the same session. It was disappointing that the lack of wifi in meeting rooms prevented many international scientists and researchers from sharing their insights.

I look forward to watching the development of PLX3397, and am sure that we will see more drug development targeting macrophages.  Coussens presentation was outstanding and the highlight of SABCS for me.

1 Comment

San Francisco – “Translational research is the key to successful drug development” according to William N. Hait MD, PhD, global therapeutic area head of oncology, Johnson & Johnson.

Hait presented a plenary session on “overcoming barriers to new drug development” at the recent AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics International Conference in San Francisco.

How do we define translational research?

The definition Hait most likes is from Duke Ellington: “if it sounds good, it is good

The challenge of drug development is that with rare exceptions the process is slow, inefficient and expensive.

Hait outlined several challenges to translational research, including:

  • Complexity – imagine blocking the traffic in mid-town manhattan. If you blocked one cross-town route, traffic would slow and then find another route.

This in my opinion is a good visual metaphor for the cross-talk that occurs in cancer. Block one target, and the cancer finds another route.  This highlights the need for combination therapy.

rational combinations of targeted agents may require studying two or more unapproved agents” said Hait.

Novel-novel combinations are something that many companies are nervous about, but if there is a solid scientific rationale then this is something I think we will see more companies doing.

For further insight into how academia is facilitating this type of combination trials, I recommend Sally Church’s interview on Pharma Strategy Blog with Gordon Mills at ECCO/ESMO in Stockholm.

  • Inaccuracy of preclinical models – our models don’t always predict preclinically what activity a drug will have in the clinic.
  • Efficacy of Clinical Trial Recruitment  – need to have alignment of incentives. 20% of US patients are eligible for clinical trials, but only 3% participate.
  • Developing Biomarkers.  Difficult to obtain serial biopsies for oncology biomarker analysis.  Circulating tumor cells may be future, but current instruments can only capture and enumerate and offer limited characterization. According to Hait, the next–generation platform will be exciting.  It will allow third parties to offer additional functionality that can be integrated with the platform.
  • Drug resistance – a nemesis that just doesn’t want to go away.
  • Overcoming the Regulatory Environment – challenges include: scientific complexity, endpoint consistency, global harmonization, companion diagnostic tests, proper comparators, equipoise.

In spite of this complexity, Hait noted the FDA approved 34 new drugs in 2011. Several cancer drugs had a short time from submission to approval and met their PDUFA target date.  “These are incredible accomplishments,” he said.

  • Market Access – Hait asked the audience: “Would you buy a Porsche 911 that only works for 20% of the people, but we don’t know if you are one of the 20%?” Healthcare authorities need to decide cost/benefit of drugs, and regulatory approval does not automatically mean a new product will be reimbursed. There may be need for future trials with health comparators, or innovative agreements where the healthcare authority only pays for those patients who respond.
  • Workforce – academic physicians may end up being segmented into three groups: master clinicians, clinical investigators, physician-scientists. This may provide better career development than the current system.

Hait offered a few suggestions for improvement:

  • Move to phenotypic screening rather than target-based screening. In vivo shRNA screening was discussed.
  • Disease based drug discovery teams – the hope is that in-depth focused teams will predict better results.
  • More intense academic-industry collaboration with a focus on complementary expertise.

The limitation of this plenary presentation was that it only offered the perspective of one senior industry professional. I would have welcomed a balance of views on the barriers to new cancer drug development, and more focused take-home solutions.

If you want to hear more on this topic, AACR have a free podcast that you can download of an interview they did with Dr Hait at the Molecular Targets meeting.

1 Comment

San Francisco – the AACR-NCI-EORTC international conference on Molecular Targets and Cancer Therapeutics kicked off last Saturday with two educational sessions, including one that I attended on “Clinical Trial Paradigms in the Era of Novel Therapies.

The session had an impressive line-up of speakers:

  • New paradigms for early-phase trials (James Doroshaw)
  • Phasing out phase III trials: How much evidence do we need if the target is clearly hit? (Jaap Verweij)
  • Development of clinical trials incorporating genomic signatures: Lessons learned? (Lisa McShane)
  • Clinical trial designs for targeted therapies (John Crowley)

James H. Doroshow, deputy director for clinical and translational research at the National Cancer Institute, started his presentation by reviewing the causes of phase II trial failure:

  • 19% Safety
  • 51% Efficacy
  • 29% Strategic

He stated that the overall success rate of recent phase II trials was 18%.

As the debate continues about whether more cancer clinical trials should be done in Phase 2, the key issue according to Doroshow remains lack of a demonstrable proof of mechanism (POM) in many drug trials. That goes hand-in-hand with a lack of molecular markers which can be used to select trial subjects.

“Lack of molecular markers with proven clinical utility follows lack of clinically-demonstrable proof of mechanism”

He provocatively asked:

Should we perform early phase trials without generating evidence supporting POM patient by patient?

His view was that to obtain POM, you need to demonstrate drug action on intended tumor target early in development, prior to expectation of efficacy.

Jaap Verweij in his presentation used the examples of crizotinib, vismodegib, vemurafenib and imatinib in GIST as examples of drugs that had:

  • functionality for a target
  • aimed at a specific population
  • availability of a selection marker.

They are the poster children of targeted therapy, and he convincingly showed that the phase 1 trials of those compounds were largely predictive of the phase 3 results.

His conclusion was that phase I trial can be considered predictive of a phase III study so long as there is a large enough sample size.

We may need to look for bigger increments which should allow us to perform smaller trials,” he said. This would allow trials that are quicker and cheaper. However, he acknowledged that it was not likely we can completely eliminate phase 3 trials particularly for combination therapies or chemotherapies.

John Crowley reviewed the different phase III trial designs, including my least favorite, the “all comer” design.  The ridaforolimus sarcoma phase 3 trial presented at ASCO this year is a good example of how an “all comer” design yielded less than stellar results, and failed to identify the subset of sarcoma patients that optimally respond.  This is the type of phase 3 trial that runs the risk of failure if there are too many non-responders in the heterogeneous patient population.  This problem can often be avoided by more rigour in phase 2 trials to identify the optimal treatment period, relevant biomarkers and subsets of patients most likely to respond.

There is a lot of interest in how to design cancer clinical trials better, bring drugs to market more quickly and more efficiently.  While I enjoyed the content of this session, I did wonder whether it would have been better presented as a roundtable with more audience interaction and engagement rather than the perspective of a few.

A webcast of this session will be available on December 8 from the American Association for Cancer Research (AACR).

The recent AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics international conference in San Francisco was an informative meeting.

What I particularly liked was the strategic overview that took place in many of the plenary sessions.

As an example, Johann de Bono, Professor of Experimental Cancer Medicine at The Institute of Cancer Research/The Royal Marsden in London highlighted the potential drug development targets based on prostate cancer biology:

  • Androgen Receptor (AR)
  • Heat Shock Proteins (Hsp)
  • Signaling: HER3, MET, IGF-1R, CCL2, IL-6, Src
  • PI3K/AKT/TOR signaling
  • PARP and BRCAness
  • Estrogen receptor (ER)
  • c-MYC & CHK1

His presentation discussed the possible therapeutic approaches, and complexity involved in developing novel targeted therapies for prostate cancer.

This is something that I expect we will hear more of at the AACR special conference on Advances in Prostate Cancer Research early next year.

In particular, de Bono discussed drug development strategies to target androgen receptor signaling, and some of the future challenges including:

  • Proving to the regulatory authorities that circulating tumor cell (CTC) count falls are a robust immediate endpoint of overall survival
  • Developing improved imaging for bone metastases

As a side note, there were several posters for cabozantinib (XL184) at the meeting (available on the Exelixis website), including preliminary research on computer-aided quantitative bone scan assessment.

However, as de Bono mentioned in his presentation, “diffusion weighted MRI shows hot spots not detected by bone scans.”

2010 and 2011 were good years for prostate cancer drugs, and with new approvals for MDV3100 and radium-223 (Alpharadin) expected, 2012 is set to be another “grand cru” year, to paraphase Bertrand Tombal.

If you were not able to make it to San Francisco for the Molecular Targets and Cancer Therapeutics conference, webcasts of many sessions will be available on the AACR site.

 

The Oncologist Journal of the Society for Translational Oncology (STO) has published a video recording on prostate cancer that is well worth watching for those with an interest in this area.

At their Sept 8, 2011 CME symposium held in Belfast, a roundtable was held entitled “Prostate Cancer: Progress & Promise.”

Moderated by Bruce A. Chabner (Mass General/Harvard), the panelists were Joe O’Sullivan (Queen’s University, Belfast), Johann De Bono (The Institute for Cancer Research) and David Waugh (Queen’s University, Belfast).

Professor de Bono in the video comments that”

“with regards to our dream of eventually treating men with prostate cancer without castrating them, which must be our ultimate goal and curing them of cancer. I think we will have to focus on for example drugs targeting ERG or ERG signaling.”

Chabner then asks the good question of whether ERG is a druggable target?

To which De Bono replies that you can drug ERG by inhibiting PARP and references a paper by the Chinnaiyan group published in the May 2011 issue of Cancer Cell.

PARP inhibition represents an interesting area of prostate cancer research.

If you would like to know more, Sally Church, PhD has written about this on Pharma Strategy Blog.  See posts on “TMPRSS2: ERG may be a more useful marker than PSA in prostate cancer” and “Personalized Therapy for Prostate Cancer – is it possible?

In the STO video, De Bono discusses why he would like to replace bone scans in prostate cancer with another imaging modality that more accurately reflects the activity of the disease. Future possibilities include use of diffusion weighted magnetic resonance imaging and novel PET tracers.

There’s also a good discussion about Alpharadin for those interested in some anecdotal commentary on experiences with it.

Another notable comment by De Bono is his belief that “taxanes work in prostate cancer primarily by targeting androgen receptor signaling.” Taxanes have typically been thought to target mitosis.

De Bono goes on to say that clinical trial data being submitted for publication shows that patients who are refractory to abiraterone, are also refractory to docetaxel when they progress on it.  The suggestion is that there may be cross resistance between abiraterone and taxanes with a subgroup of patients who just don’t do well on androgen receptor (AR) targeting drugs.  The reason for this isn’t yet clear.

A new phase 2 clinical trial is starting soon that will look at the sequencing of abiraterone and cabazitaxel.  One group will receive abiraterone followed by cabazitaxel, the other cabazitaxel followed by abiraterone.

The Belfast STO symposium was the second in a three part series. The next one will be held during ASCO GU in San Francisco next year.

Another potentially useful meeting in this area is the February 2012 AACR workshop on “Advances in Prostate Cancer Research” chaired by Arul Chinnaiyan & Charles Sawyers.

Prostate cancer remains an exciting therapeutic area to watch with tremendous progress and promise of late.

Biotech Strategy Blog is 1 today!  I can’t believe that a year has gone by so quickly!  Before moving on to year 2, I thought a brief review might be interesting.

What have been the top posts on Biotech Strategy Blog this past year?

In terms of total visitors per post:

  1. Results from NEJM Lucentis v Avastin AMD CATT clinical trial
  2. AUA Results from PIVOT study show no benefit from radical prostatectomy in low risk early stage patients
  3. ASCO 2011 Cabozantinib (XL184) may be an exciting new prostate cancer drug
  4. Merck’s capthepsin-K inhibitor odanacatib in osteoporosis
  5. Update from AACR on new prostate cancer drugs to watch

For those who like metrics:

  • Highest number of reads per month was in May (19,927)
  • Year to date there have been 79,179 visitors
  • Most visited day was September 22, 2011 (2136 reads)

What have been some of the other posts that I enjoyed writing about?

My top 5 (not in rank order) would be:

  1. Alpharadin will be new treatment option for prostate cancer
  2. Patient advocacy session at European Hematology Assocation EHA Congress shows impact of drug adherence on outcome
  3. How nanotechnology may revolutionize the detection of traumatic brain injury using a sensor that changes color
  4. Innovation in Nanotechnology will lead to improved drug delivery, diagnostics & imaging
  5. Insights of the decade

Finally, I have produced 4 videos that you can watch on the biotechstrategy channel on YouTube.

http://youtu.be/nDvY7opm3Fs

http://youtu.be/_oAJ1fU0PT4

http://youtu.be/hM_wmjaqDyc

http://youtu.be/i5GNBmuISqQ
It’s been a busy but enjoyable year. Biotech Strategy Blog is still a work in progress.  If you have enjoyed a particular series of posts or would like me explore a topic or theme in the future, do email me or post a comment.

error: Content is protected !!