Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts by MaverickNY

This morning my emails greeted me with an announcement that the long awaited phase III SUCCEED trial data in soft tissue sarcomas from Ariad and Merck has been announced:

“… showing that ridaforolimus, an investigational oral mTOR inhibitor, met the primary endpoint of improved progression-free survival (PFS) compared to placebo in the Phase 3 SUCCEED trial conducted in patients with metastatic soft-tissue or bone sarcomas who previously had a favorable response to chemotherapy.

Merck is currently developing ridaforolimus in multiple cancer indications under an exclusive license and collaboration agreement with ARIAD.   Complete findings from the SUCCEED trial will be submitted for presentation at an upcoming medical meeting this year.”

In the analyst teleconference I just listened to, the CEO Harvey Berger also stated that aside from presenting the data at a meeting this year, Merck was planning on filing the data with the regulatory authorities.

The good news is that no new safety signals have emerged and the common side effects reported previously, ie stomatitis (mouth sores), fatigue, diarrhea and thrombocytopenia appear unchanged.

Correspondence and interviews with a number of the SUCCEED trialists in the US and EU over the last year has taught me that ridaforolimus is generally well tolerated and quite a few patients have experienced prolonged stable disease (SD) while on ridaforolimus maintenance therapy between chemotherapy.

According to Ariad, the initial aggregated data from SUCCEED was as follows:

“Based on the full analysis of 552 PFS events in 711 patients, determined by an independent review committee, the blinded prospective study achieved its primary endpoint, with a statistically significant (p=0.0001) 28 percent reduction by ridaforolimus in the risk of progression compared to placebo (hazard ratio=0.72). Determination of median PFS for each arm of the trial demonstrated that ridaforolimus treatment resulted in a statistically significant 21 percent (3.1 week) improvement in median PFS (ridaforolimus, 17.7 weeks vs. placebo, 14.6 weeks).

Based on the full analysis of PFS determined by the investigative sites, there also was a statistically significant (p<0.0001) 31 percent reduction by ridaforolimus in the risk of progression compared to placebo (hazard ratio=0.69).  Ridaforolimus treatment resulted in a statistically significant 52 percent (7.7 week) improvement in median PFS (ridaforolimus, 22.4 weeks vs. placebo, 14.7 weeks).”

There are several important things to note here:

  1. The PFS is less than a month, but highly significant
  2. The conditions for the SPA (primary endpoint was PFS) appear to have been met
  3. The last patient enrolled early last year, so hopefully the OS data (secondary endpoint) will be ready by the filing and will be positive
  4. The excellent hazard ratios suggest that the curves are well separated, leading me to think/hope that the OS might actually be more encouraging than PFS
  5. PFS is a difficult surrogate end point to measure; many trials end up with a difference between investigator estimate and central review as a result

We also need to look at these results in the context of soft tissue sarcomas (STS) as a disease.  By this, I’m thinking about the following issues:

  • STS is a heterogeneous disease made up of many different subtypes
  • It is a difficult to treat, aggressive disease and many patients are treated with multiple lines of chemotherapy to progression but sadly still relapse as resistance develops
  • Some subtypes are highly chemosensitive, some are not
  • Stable disease and quality of life are valuable to people with sarcoma
  • The danger of a ‘catch-all’ trial is that the non-responders cancel out the responders, lowering the overall response rate
  • Did any particular subsets do better on ridaforolimus than others?
  • Did any biomarkers emerge around mTOR, PI3-kinase, AKT, MEK or some other factor?
  • What factors were associated with the development of resistance?
  • If so, would this help us learn what might be a useful combination approach going forward?

Overall, based on what I’ve seen, this data looks promising so far and I’m keen to see more granular data when it is presented at a scientific meeting soon… Perhaps the data made the ASCO late breaking abstract cut-off this month?

7 Comments

PI3-kinase inhibition is becoming quite a hot topic in cancer research – for those of you interested in learning about the compounds in this area, you can find out more from previous posts here and here as background information before reading the summary of the latest research below.

A friend kindly alerted me to a new article by Donev et al., (2011) that discussed how PI3K inhibition can potentially overcome resistance associated with EGFR tyrosine kinase inhibitors (TKIs) in EGFR mutated lung cancer.  This is an important area in both cancer research and in the clinic, because while erlotinib and gefitinib have been approved for the treatment of lung cancers where the EGFR mutation is present, efficacy is usually limited by the development of drug resistance.

There are a number of reasons for the development of resistance with EGFR inhibitors:

  1. Pao et al., and Kobayashi et al., (2005) demonstrated that acquired resistance can be due to a second mutation in the EGFR kinase domain, ie T790M.
  2. Engleman et al., (2007) observed that MET gene amplification is also an important mechanism for driving resistance by activating ERBB3 Signaling.
  3. Yano et al., (2008) demonstrated the importance of hepatocyte growth factor (HGF) inducing gefitinib resistance of lung adenocarcinoma with EGFR-activating mutations by activating MET, which restores phosphorylation of downstream MAPK-ERK1/2 and PI3K-Akt pathways.

The role of MET and HGF in EGFR mutations is an interesting one.  Turke et al., (2010) subsequently demonstrated that MET activation by its ligand, HGF, also induces drug resistance through GAB1 signaling.  HGF accelerated the development of MET amplification both in vitro and in vivo.  In addition, EGFR resistance, due to either MET amplification or autocrine HGF production, was “cured in vivo by combined EGFR and MET inhibition.” Now, I don’t normally like the use of the word ‘cure’ but it did get my attention!  What was clear from their data was that rare MET-amplified cells exist in some EGFR-mutant lung cancers before, treatment.  This suggests that HGF is able to accelerate MET amplification by expanding the preexisting MET-amplified cells.

Kasahara et al., (2010) did some research looking at serum HGF and found that it was strongly related to the outcome of EGFR-TKIs treatment. It would therefore be interesting to develop the idea further to determine if the approach could be used to refine the selection of patients expected to respond to EGFR-TKIs treatment.

A more recent paper by Donev et al., (2011) takes the HGF resistance idea further.  They demonstrated that transient blockade of PI3K-Akt pathway by PI3K inhibitor and gefitinib could overcome HGF-mediated resistance to EGFR-TKIs by inducing apoptosis in both in vitro and in vivo models.  This approach may be particularly useful in those patients who develop acquired resistance to EGFR therapy driven by HGF.

Impact of the translational research findings:

Of course, we don’t yet know how all this body of research will translate into the clinic in humans, but these recent findings do begin to give us some pointers of where to begin. Based on the research to date,  I don’t think it’s going to be as simple as adding a PI3K or MET inhibitor into the mix with an EGFR TKI in every patient though.

A logical and smart approach would be to categorise patients into several groups and determine treatment according to the tumour characteristics:

  • those who are MET amplified upfront (i.e. MET high vs MET low)
  • those with a T790M mutation present or develop one during treatment
  • those who develop HGF-acquired resistance

There are some T790M inhibitors in very early development, but most of these seem to have languished in pipelines to date.   I do recall that Pfizer had one in development (PF00299804), based on a paper by Engleman et al., (2007).  However, I have not heard much about it lately, probably because they have a large oncology pipeline and are focusing on getting crizotinib to market in advanced lung cancer with ALK translocations.

Boehringer have pan ERB agent in the clinic, BIBW 2992, in newly diagnosed patients as a single agent, but it is too early to tell how effective it will be.  Sometimes being first into the clinic isn’t always the best approach, because others slightly behind can take advantage of new research findings and design smart combination trials based on the translational findings.

MET inhibitors are much more common and a number of pharma companies are looking at these in phase II or III trials in combination with erlotinib in lung cancer.  The most advanced are ArQule’s ARQ-197 and Roche’s MetMAB, both of which had data presented at ESMO last autumn.  Roche showed clear differences in response to MetMAB in patients segmented for MET high vs MET low expression.  In contrast, ArQule reported the data in the aggregate at ESMO, so it wasn’t as easy to see what was happening from a biomarker perspective.  They also announced that they have accrued the first patient in their phase III trial earlier this month so it will be a little while before we hear how successful these two compounds will be.

PI3-kinase inhibitors are much more plentiful and include BEZ-235 (Novartis) and GDC-0941 (Roche) amongst many others, some of which I previously discussed last year.   These may have a role to play in HGF-mediated EGFR resistance, for example, where the current translational data suggests that adding a PI3K inhibitor may be an effective strategy to pursue.

Meanwhile, on the HGF front, Okamoto et al., (2010) reported that preclinical results with TAK-701 (Takeda/Millennium), a humanized monoclonal antibody to HGF. They found that it reversed gefitinib resistance induced by tumor-derived HGF in non-small cell lung cancer (NSCLC) with an EGFR mutation in a human lung cancer cell model. These results are encouraging and give some clues of where to start in the clinic with this compound in combination with an EGFR inhibitor.

Overall, this field is looking very promising, but it will be very important to carefully translate the translational research findings into different patient subsets in clinical trials associated with EGFR mutated NSCLC and EGFR TKIs.

References:

ResearchBlogging.org

Pao, W., Miller, V., Politi, K., Riely, G., Somwar, R., Zakowski, M., Kris, M., & Varmus, H. (2005). Acquired Resistance of Lung Adenocarcinomas to Gefitinib or Erlotinib Is Associated with a Second Mutation in the EGFR Kinase Domain PLoS Medicine, 2 (3) DOI: 10.1371/journal.pmed.0020073

Kobayashi, S., Boggon, T., Dayaram, T., Jänne, P., Kocher, O., Meyerson, M., Johnson, B., Eck, M., Tenen, D., & Halmos, B. (2005).
Mutation and Resistance of Non–Small-Cell Lung Cancer to Gefitinib
New England Journal of Medicine, 352 (8), 786-792 DOI: 10.1056/NEJMoa044238

Engelman, J., Zejnullahu, K., Mitsudomi, T., Song, Y., Hyland, C., Park, J., Lindeman, N., Gale, C., Zhao, X., Christensen, J., Kosaka, T., Holmes, A., Rogers, A., Cappuzzo, F., Mok, T., Lee, C., Johnson, B., Cantley, L., & Janne, P. (2007). MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling Science, 316 (5827), 1039-1043 DOI: 10.1126/science.1141478

Yano, S., Wang, W., Li, Q., Matsumoto, K., Sakurama, H., Nakamura, T., Ogino, H., Kakiuchi, S., Hanibuchi, M., Nishioka, Y., Uehara, H., Mitsudomi, T., Yatabe, Y., Nakamura, T., & Sone, S. (2008). Hepatocyte Growth Factor Induces Gefitinib Resistance of Lung Adenocarcinoma with Epidermal Growth Factor Receptor-Activating Mutations Cancer Research, 68 (22), 9479-9487 DOI: 10.1158/0008-5472.CAN-08-1643

Turke, A., Zejnullahu, K., Wu, Y., Song, Y., Dias-Santagata, D., Lifshits, E., Toschi, L., Rogers, A., Mok, T., & Sequist, L. (2010). Preexistence and Clonal Selection of MET Amplification in EGFR Mutant NSCLC Cancer Cell, 17 (1), 77-88 DOI: 10.1016/j.ccr.2009.11.022

Kasahara, K., Arao, T., Sakai, K., Matsumoto, K., Sakai, A., Kimura, H., Sone, T., Horiike, A., Nishio, M., Ohira, T., Ikeda, N., Yamanaka, T., Saijo, N., & Nishio, K. (2010). Impact of Serum Hepatocyte Growth Factor on Treatment Response to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Non-Small Cell Lung Adenocarcinoma Clinical Cancer Research, 16 (18), 4616-4624 DOI: 10.1158/1078-0432.CCR-10-0383

Okamoto, W., Okamoto, I., Tanaka, K., Hatashita, E., Yamada, Y., Kuwata, K., Yamaguchi, H., Arao, T., Nishio, K., Fukuoka, M., Janne, P., & Nakagawa, K. (2010). TAK-701, a Humanized Monoclonal Antibody to Hepatocyte Growth Factor, Reverses Gefitinib Resistance Induced by Tumor-Derived HGF in Non-Small Cell Lung Cancer with an EGFR Mutation Molecular Cancer Therapeutics, 9 (10), 2785-2792 DOI: 10.1158/1535-7163.MCT-10-0481

Donev, I., Wang, W., Yamada, T., Li, Q., Takeuchi, S., Matsumoto, K., Yamori, T., Nishioka, Y., Sone, S., & Yano, S. (2011). Transient PI3K Inhibition Induces Apoptosis and Overcomes HGF-mediated Resistance to EGFR-TKIs in EGFR Mutant Lung Cancer Clinical Cancer Research DOI: 10.1158/1078-0432.CCR-10-1993

Engelman, J., Zejnullahu, K., Gale, C., Lifshits, E., Gonzales, A., Shimamura, T., Zhao, F., Vincent, P., Naumov, G., Bradner, J., Althaus, I., Gandhi, L., Shapiro, G., Nelson, J., Heymach, J., Meyerson, M., Wong, K., & Janne, P. (2007). PF00299804, an Irreversible Pan-ERBB Inhibitor, Is Effective in Lung Cancer Models with EGFR and ERBB2 Mutations that Are Resistant to Gefitinib Cancer Research, 67 (24), 11924-11932 DOI: 10.1158/0008-5472.CAN-07-1885

7 Comments

There’s been a lot of noise on the internet from some early adopters who think Really Simple Syndication (RSS) is dead with so much information now available, often shared by people through Twitter and Facebook.  For me, though, that’s not true and here’s why:

  1. It depends on the tool you’re using to consume the RSS feeds
  2. Not many people in my circle share cancer journal items I’m interested in
  3. I use RSS feeds as a searchable database for key information

What tool is useful?

One tool I particularly love is Reeder, which can be used on a Mac computer, an iPad, iPhone or Android device essentially to read your RSS feeds hosted in Google Reader. You can read them online or offline.  This app allows you to skim through the news or science items in a more user friendly way.

Instead of clicking j/k and taking ages to move through items, the layout is much more pleasing and easier to read, like this:

Using RSS to search for interesting or relevant articles:

One of the challenges though, is that some journals such as Nature (in the example above) only provide the title and authors without even an abstract, which is frustrating and more than a little ungenerous of the publisher.

Others such as the Blood journal, however, do provide a useful summary of an article.  I found this one by quickly searching for ROS signaling, for example:

Once, found, it’s easy to star or bookmark for later use.  You can also cut/paste the information or quotes easily to notes or a presentation, adding to the overall utility of the tool.  It’s also a convenient way to search for information, assuming you have input journal RSS feeds into Google Reader, as I have done.  Although time consuming to do, it is well worth the effort in the long run.

Pictures also show up really well in Reeder:

I particularly enjoy skimming, searching and bookmarking my journal RSS feeds while offline on a plane on a laptop as it is a great productivity tool, but this approach would work equally well on an iPad too, which would make flipping through the curated material even more user friendly.

If you have a Mac, iPhone, iPad or Android gadget, then head over here to check it out.

What do you like about RSS?

3 Comments

At the recent International Society of Gastroenterological Carcinogenesis (ISGC) meeting hosted by MD Anderson Cancer Centre that I attended in Houston, one of the topics mentioned the potential role of Fibroblast Growth Factor Receptors (FGFR) in carcinogenesis. I thought this was a great opportunity to research the area further.

A recent review of the role of FGFR in carcinogenesis fortuitiously appeared in Molecular Cancer Research:

“The fibroblast growth factor receptors (FGFR) play essential roles both during development and in the adult.  Upon ligand binding, FGFRs induce intracellular signaling networks that tightly regulate key biological processes, such as cell proliferation, survival, migration, and differentiation. Deregulation of FGFR signaling can thus alter tissue homeostasis and has been associated with several developmental syndromes as well as with many types of cancer.

In human cancer, FGFRs have been found to be deregulated by multiple mechanisms, including aberrant expression, mutations, chromosomal rearrangements, and amplifications.”

They also went on to define what carcinogenesis is based on what we learned from Hanahan and Weinberg (2000) in their classic paper, The hallmark of Cancer:

“Carcinogenesis is a multistep process during which normal cells are transformed into cancer cells by accumulating several genetic changes and acquiring several common features that promote the malignant phenotype, often referred to as the hallmarks of cancer.

The six classic hallmarks of cancer include self-sufficiency in growth signals, insensitivity to antigrowth signals, limitless replication, evasion of apoptosis, sustained angiogenesis, and the ability to invade tissue and form metastasis.”

Bearing in mind that Hanrahan and Weinberg’s paper was published over a decade ago, there were some interesting observations that hold true:

“It is increasingly apparent that the growth deregulation within a tumor can only be explained once we understand the contributions of the ancillary cells present in a tumor—the apparently normal bystanders such as fibroblasts and endothelial cells—which must play key roles in driving tumor cell proliferation.”

Their article is now open access, so you can see how they described the role of these cells with cancer cells.

Going back to Haugsten et al., (2010) review, we learn new developments in carcinogenesis that have taken place over the last decade relating to FGFR, which has received much less attention than other receptors such as VEGF, PDGF, EGFR and IGF-1R as a potential target.   The FGFR family consists of four genes encoding the tyrosine kinase receptors (FGFR1 to FGFR4).  Downstream, the pathway becomes quite complex, so it will be interesting to see which factors emerge as escape routes through cross-talk and feedback loops.

FGFR pathway in Cancer

The potential role of FGFR in cancer carcinogenesis is summarised in the table below:

FGFR in Cancer

There are a variety of different cancers potentially affected, but FGFR is not always overexpressed where it is amplified and may not always be contained in the amplification.  Sometimes overexpression merely indicates a poorer prognosis or the development of resistance.

The chances of it being a drugable target and therefore more likely to have a meaningful clinical impact is probably greater where there are mutations or fusion proteins, since these often (but not always) have aberrant activity associated with them.  The challenge is therefore figuring where it might be a passenger or a driver gene.

Turner and Grosse (2010) also reviewed the FGFR pathway as a new area of research:

“Although FGF signalling can drive tumorigenesis, in different contexts FGF signalling can mediate tumour protective functions.  The identification of the mechanisms that underlie these differential effects will be important to understand how FGF signalling can be most appropriately therapeutically targeted.”

Obviously, we won’t know the answer until clinical trials with the small molecule inhibitors and monoclonal antibodies are completed, but it certainly looks to be a worthwhile area of exploration.  Hynes and Dey (2010) discussed the potential role in breast cancer in more detail, noting the findings of Roidl et al (2009):

“In breast cancer cell lines, it has been reported that increased levels of FGFR4 are found in cells resistant to chemotherapeutics.”

This suggests a combination strategy in those cases may be worthwhile.

For those who missed it, I also posted about the potential role of FGFR1 in lung cancer last month, which also included some of the inhibitors emerging in this class.

References:

ResearchBlogging.orgHaugsten, E., Wiedlocha, A., Olsnes, S., & Wesche, J. (2010). Roles of Fibroblast Growth Factor Receptors in Carcinogenesis Molecular Cancer Research, 8 (11), 1439-1452 DOI: 10.1158/1541-7786.MCR-10-0168

Hanahan, D., & Weinberg, R. (2000). The Hallmarks of Cancer Cell, 100 (1), 57-70 DOI: 10.1016/S0092-8674(00)81683-9

Turner, N., & Grose, R. (2010). Fibroblast growth factor signalling: from development to cancer Nature Reviews Cancer, 10 (2), 116-129 DOI: 10.1038/nrc2780

Hynes, N., & Dey, J. (2010). Potential for Targeting the Fibroblast Growth Factor Receptors in Breast Cancer Cancer Research, 70 (13), 5199-5202 DOI: 10.1158/0008-5472.CAN-10-0918

Roidl A, Berger HJ, Kumar S, Bange J, Knyazev P, & Ullrich A (2009). Resistance to chemotherapy is associated with fibroblast growth factor receptor 4 up-regulation. Clinical cancer research : an official journal of the American Association for Cancer Research, 15 (6), 2058-66 PMID: 19240166

2 Comments

SpringPad is a new free tool I’ve recently been playing with and it has quickly become a way to sort and organise information easily. In the past, I’ve been a big Evernote fan, but while it is useful for collecting abstracts, photos, papers etc, SpringPad has a whole different set of utilities that I find myself using on a daily basis, both in the office and while mobile at conferences.

One of the challenges in my work is sifting through vast amounts of data and generating useful insights, either for posts here on this blog or as a consultant. I’ve tried a lot of different web 2.0 tools over the last 7 years but occasionally one comes along that sticks in my workflow. SpringPad is one of them.

The first thing I did after signing up was download the iPhone app and the web clipper for Chrome, my preferred browser. This makes life a lot easier when you come across anything interesting:

SpringPad Web Clipper

You can also email items such as webpages, links and PDF files to SpringPad.

Once in the SpringPad web app (similar layouts for the iPhone are available in an app too), your top level notebooks appear something like this:

SpringPad Notebooks

You can colour code them for easy visual appeal and finding things. I’ve also hidden the mission critical client projects, but you can see the general gist of what my recent science topics looks like.

The nice thing about this approach is that you can create Notebooks by topics and then once you’ve clipped or emailed relevant information to SpringPad, it can be organised efficiently.

For example, the JP Morgan Healthcare conference is ongoing this week with lots of useful snippets emerging by company, drug and pathway.   I can clip, then tag the information and also assign it to several Notebooks.  Information emerging from the meeting on Keryx’s perifosine might get added to the Keryx, colorectal cancer and myeloma Notebooks, for example.  This makes it easier to find information later when you search for it, or later look at all the information you have collected to date on say, lung cancer or a particular pathway, to look at the big picture trends. I also diligently tag items across a broad range of topics so they will appear later in the database searches.

Another useful feature of SpringPad is that you can collate information around an event.

Once inside a notebook, for example, the recent one I created for the ISGC meeting at MD Anderson to keep me organised with a one stop shop for all the preconference information, to-dos and post conference notes looked like this:

SpringPad Notebook

While travelling to this meeting, I had everything I needed for the event in the iPhone app and could add new notes, to dos, places, contacts etc while on the road for other projects. This worked really well, even on the plane, since SpringPad will sync the notes once internet access is available later.

The iPhone is small so it is not good for rapidly note taking at meetings and wifi was gippy at best, so I made most of the notes on my Mac laptop in Twitter using a hashtag and also in an offline text app, Notational Velocity, which syncs with Simplenote. I’m now looking to see if I can email my notes on each presentation to SpringPad or worst case scenario, cut/paste them into the notes created. Another way to do this efficiently would be to use an iPad, but that’s still on my wish list in the Gear Notebook 🙂

Assigning dates to To-Dos and items is a really useful feature – you can check your Alert box and see what’s immediately due.

Another feature I like is the ability to import Delicious bookmarks (I have 3,000 of them!) as well as the associated tags, so these are now searchable in the context of any other information I might have collated in SpringPad. When a client rings up or sends an email asking about something, this makes the scientific, commercial or clinical answer much easier to find than Googling and getting lots of spammy results, which seems de rigeur in public searches of late.

There’s a lot more functionality in SpringPad not covered in this review, but I will add more updates as it becomes more familiar and a bigger database is built up. Has anyone else tried SpringPad yet? If so, what were your experiences or do you have any cool tips to share?

9 Comments

My colleague who writes the Biotech Strategy Blog, posted an interesting analysis this morning on the Matrixx Zicam case currently being reviewed by the United States Supreme Court (SCOTUS).  At the heart of the issue is whether or not adverse events should be disclosed to investors, even if not statistically significant. The way the questioning appears to be going from the Justices so far, it looks like they believe they should be.

The reason I bring this topic up is because it has important implications for Pharma and Biotech companies.

Recently, several obesity drugs were reviewed by the FDA Advisory Committees.  It’s not an area I usually follow, except that one of them, Arena Pharmaceuticals ($ARNA), caused a stir when many analysts and investors were surprised by the FDA briefing documents.  In it, was a hidden gem – the drug in question appeared to have an increased risk of developing cancer in rats – but the company hadn’t publicly revealed this fact. Why?  Because the CEO, Jack Lief, and the Board didn’t think it was material.

Inevitably, a lot of investors were rather dismayed not to have known that information before deciding whether or not to invest in the company.  Most of the industry watchers on Twitter, including myself, were certainly a little flabbergasted to hear it for the first time from the FDA.

Of course, the drug did not get recommended for approval, partly because of the marginal efficacy data, but also there was genuine concern over heart valve problems, and it is hard to imagine that the undeclared rat cancer didn’t factor in either.  In the end, a complete response letter (CRL) was issued.

I love Adam Feuerstein of The Street’s tweet around that time:

Adam Feuerstein ARNA tweet on rat cancer

Now, if we fast forward to the SCOTUS discussion on this very issue, it is not hard to see that this kind of information ought to be revealed to allow investors to make a rational decision on whether to invest or not.

The final SCOTUS decision is expected by the summer, so it will be interesting to see what happens if they decide that adverse events, whether significant or not, should be declared.  personally, I think they should and people can then do their own due diligence and research on whether they think it increased their risk tolerance or not.

This decision may well have a far reaching impact on the Pharma and Biotech industry and force company Boards to be far more rigorous and diligent in providing relevant information to potential investors.

4 Comments

Iniparib phase II data in NEJM

At ESMO last summer the initial phase II trial data on a PARP inhibitor, iniparib (sanofi-aventis), was presented in triple negative breast cancer (TNBC). Unfortunately, I missed that session as it clashed with something else I wanted to see, but the data has now been published in the New England Journal of Medicine.

The references are provided below for the link to the online article and accompanying editorial (subscription required).

The front page of the journal provides a public link to the key overall survival data so far. As you can see, the bottom line is that the Kaplan-Meier curves do not cross over and a clear benefit in favour of the iniparib plus chemotherapy over chemotherapy alone is shown below:

Iniparib Phase II survival data

Source: NEJM

The primary endpoint of the trial was progression-free survival (PFS). The median PFS in the iniparib arm was 5.9 months compared with 3.6 months in the chemotherapy arm. Overall survival (OS) was a secondary endpoint.  This data (in the K-M curve above) showed that the OS in the iniparib arm was 12.3 months compared with 7.7 months among those who received chemotherapy alone.  This translates to a 43% relative reduction in the risk of death.

Looking at the adverse event data closely, iniparib looks to be a reasonably well tolerated drug.   The most common side effects in the iniparib group included neutropenia, anemia, thrombocytopenia, fatigue/asthenia, nausea and constipation.  With regards to grade 3/4 adverse events, the most commonly reported ones in the iniparib group were neutropenia, anemia, thrombocytopenia, leukopenia and fatigue/asthenia. There were two fatal adverse events (3.4%) in the chemotherapy-alone arm and three (5.3%) in the iniparib arm, all attributed to disease progression within 30 days of receiving study treatment.

According to the company (sanofi-aventis), the phase III trial in women with triple negative breast cancer is already ongoing and results are expected in 2011.  Assuming these results are reproducible and no additional safety signals emerge, we can expect the data to be filed for approval with the EMA and FDA.

What does all this data mean?

To put this class in context, I first blogged about PARP inhibitors in 2006, before they became a hot topic and have followed their progress since (just type PARP in the search box to the right to see the other posts over time).  Several others have since joined the clinical pipelines including olaparib (AstraZeneca) and veliparib (Abbott).  What has been interesting from a strategy perspective, is that both olaparib and veliparib have focused largely on BRCA1 and 2 mutated cancers, whereas iniparib has been studied in triple negative breast cancer, which largely consists of basal-type rather than luminal histology. Of relevance here is that most BRCA1 breast cancers also have basal histology.  Together, we could consider them both to have “BRCAness” as many breast cancer specialists have begun to refer them.

The latest article published last week was accompanied by a hard hitting editorial from Drs Carey and Sharpless (UNC).   They noted several weaknesses in the current trial of iniparib, namely:

“The cohort was small, the end points were assessed by the investigators, the gemcitabine–carboplatin regimen is unconventional, and there were imbalances at baseline in prognostically important characteristics favoring the iniparib group.

We cannot tell whether the benefit from the PARP inhibitor accrued to all triple-negative tumors equally or whether the benefit preferentially accrued to a subgroup of BRCA-deficient tumors, with less effect in those without the deficiency.”

In other words, did a subset of women with TNBC and BRCA1 or 2 do better than those without?  It’s impossible to tell from the data presented to date and would be a most instructive subset analysis.

Many of us will wonder about biomarkers too, and whether something will emerge from the trials that indicates a way to predict which women might best respond to the therapy rather than treat everyone with the disease to achieve a 56% clinical benefit.  Could the other 44% be spared of the systemic effects of a therapy that is unlikely to work for them?  Part of me is resigned to thinking this is a classic chemotherapy approach where you add in a new agent to standard chemo and hope that it moves the needle enough to prove significant.  What we really need is a more detailed analysis with targeted therapies that looks at responders and non-responders to determine the underlying mechanisms for greater predictive or prognostic sensitivity.

The editorial also went on to state:

“In addition, unlike with iniparib, it has been challenging to combine several of these other agents with chemotherapy. Iniparib is a much less potent inhibitor of PARP1 (with approximately 0.1% the potency) than most other agents of this class.

The present study does not include a pharmacodynamic assessment of PARP activity in the patients receiving iniparib, and it is unclear whether the therapeutic efficacy of this agent correlates with PARP inhibition in these patients.

Therefore, the low potency, reduced toxicity when combined with chemotherapy, and possible BRCA-independent activity of iniparib distinguish it from other members of the class; at least part of its antitumor efficacy may be independent of PARP inhibition.”

In other words, it may actually be a PARP-like inhibitor rather than a pure PARP inhibitor.  Clearly we have much to learn about PARP inhibitors and iniparib in particular, but hopefully some more useful granular data analysis will evolve from the phase III trial.

Obviously, we will have to wait for the confirmatory phase III results due next year, but if they are are positive and should iniparib receive approval, it may well be the first major advance for women with this type of breast cancer, who currently have a poorer prognosis than those with ER/PR+ disease.  That’s good news for women with triple negative disease and something to be hopeful about.

References:

ResearchBlogging.org O’Shaughnessy, J., Osborne, C., Pippen, J., Yoffe, M., Patt, D., Rocha, C., Koo, I., Sherman, B., & Bradley, C. (2011). Iniparib plus Chemotherapy in Metastatic Triple-Negative Breast Cancer New England Journal of Medicine DOI: 10.1056/NEJMoa1011418

Carey, L., & Sharpless, N. (2011). PARP and Cancer — If It’s Broke, Don’t Fix It New England Journal of Medicine DOI: 10.1056/NEJMe1012546

3 Comments

I am always trying to find novel ways to use social media and new marketing channels to engage blog readers and distribute content, so for the princely sum of $0.99 a month you can now read Pharma Strategy Blog on your Kindle.

If you have a Kindle and would like the convenience of reading Pharma Strategy Blog offline at your leisure, on a bus or airplane, I hope you will think about signing up. Amazon offer a 14 day free trial.

Please click on the link below or on the right hand side sidebar to subscribe. I look forward to hearing from you as to whether this proves to be a successful way to deliver content.

On Thursday this week I’m off to the GI Carcinogenesis meeting hosted by MD Anderson Cancer Center, you can find out more about the event here.

It’s a brand new meeting for me, but according to the program:

“The ISGC is comprised of basic, translational and clinical scientists.  This conference will encourage and develop research and communication in the areas of gastroenterological biology and oncology in both basic and clinical aspects through joint meetings with international and national gastroenterologists.”

I’m particularly looking forward to hearing what Lee Ellis has to say on cancer stem cells and the microenvironment, as well as Emanuel Petricoin on molecular profiling in GI cancers.  There are a whole host of other really interesting talks too, as you can see from the program agenda.

When I first looked at the faculty, my initial reaction was, “Oh my!” It’s a quite serious line-up of some of the top GI cancer researchers and certainly not easy to get them all in the same place together, so it will be fun to chat with them in the poster sessions and get their perspective on the latest happenings in this field.

The meeting runs through Saturday, so I’ll try and post a daily synopsis, as time permits.

If I hadn’t been following Dr Raymond DuBois, the MD Anderson Provost and Co-Chair of the meeting on Twitter, I would have missed this altogether – the power of social media in spreading and communicating awareness of these special events is very much here to stay.

If you have any burning questions in this area, please do add them in the comments below and I will do my best to ferret out some answers.

1 Comment
error: Content is protected !!