Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts by MaverickNY

Yesterday, I covered some of the key pathways and kinases associated with cell energy metabolism, LKB1 and AMPK.  These, together with Insulin-like Growth Factor-I (IGF-I) and the insulin receptor (IR), appear to play important roles in the broader regulation of energy and homeostasis.  Experimental evidence suggests that an overexpression of IGF-I is implicated in pancreatic tumours, for example. Increased IGF-II and decreased IGF binding protein (IGFBP)-3 serum concentrations have also been linked to a number of other cancers (see journal link below).

If we look at the IGF-IR pathway, we can see more clearly how they all interlink and how mTOR, LKB1 and AMPK may all be a critical part of the process:

IGF1RSource: Tao et al., (2007)

Research conducted over the past two decades has shown the importance of the type 1 insulin-like growth factor receptor (IGF-1R) in tumorigenesis, metastasis, and resistance to existing forms of cancer therapy. We also now know that feedback and cross-talk between IGF1R and IR can exist, driving hyperglycemia and free insulin production, as shown in a previous post regarding IGF-1R inhibition with figitumumab.  

Clearly, there are drugs commercially available that reduce hyperglycemia in diabetes, so the next logical step would be to see what happens if they were to be used in cancer patients or people with a very high risk for developing cancer.

Background:

The journal Cancer Prevention Research has just published an interesting series of research papers around metformin, a generically available oral biguanide for the treatment of Type II diabetes, in cancer prevention.  An excellent overview to the topic was covered in a comprehensive review of metformin for oncology applications by Michael Pollak, which is well worth reading.

In short, metformin was originally derived from a plant extract from the French lilac and activates AMPK in the liver.  This means that it plays an important role in insulin signaling, whole body energy balance, and the metabolism of glucose.  In diabetes, metformin therefore improves hyperglycemia primarily through its suppression of hepatic glucose production, ie gluconeogenesis. 

If we accept the research that shows high levels of free IGFI (from the higher IGF-I/IGFBP-3 molar ratio) are important in cancer growth, then it makes logical sense to see what effects taking metformin might have on cancer prevention and risk reduction in animal models and humans.

Effect of metformin on lung cancer

Memmot et al., published a paper entitled, "Metformin Prevents Tobacco Carcinogen– Induced Lung Tumorigenesis" in Cancer Prevention and Research. The idea was that activation of the mammalian target of rapamycin (mTOR) pathway is an important and early event in tobacco carcinogen–induced lung tumorigenesis, thus therapies that target mTOR might be effective in the prevention or treatment of lung cancer.  Since metformin activates AMPK, which in turn inhibits mTOR, they decided to investigate the possibilities in a mouse model.  The mice were given a known cancer causing carcinogen, nitrosame ketone (NNK) and a group were treated with metformin and compared to controls (no treatment).

What they found was startling:

"Oral administration of 1 or 5 mg/mL metformin decreased lung tumor burden in mice by 38% and 53%, respectively."

What happened to the control mice who did not receive metformin? 100% of them developed tumorigenesis.  To put these findings into perspective:

"The steady-state levels of metformin in mice given 5 mg/mL are similar to those in diabetic patients using metformin, suggesting the possibility that clinical prevention of lung cancer could be achieved with standard oral dosing."

Unsurprisingly even greater results were observed with direct intravenous metformin:

"intraperitoneal administration of metformin was more effective than oral administration and decreased tumor burden by 72%."

Overall, the researchers found that inhibition of the mTOR pathway in tumours was associated with decreases in levels of circulating IGF-I and insulin, which may well explain the dramatic results they saw with metformin.

Effect of metformin on colorectal cancer

Animal models are all very well, but what about human data? Japanese researchers have now reported the first study of metformin in people without diabetes, albeit on a small scale. Hosono et al., published an article, "Metformin Suppresses Colorectal Aberrant Crypt Foci in a Short-term Clinical Trial" after their earlier work in rodents. Rectal aberrant crypt foci (ACF) are an endoscopic surrogate marker of colorectal cancer, essentially an early precursor to malignant disease.

This work looked at prospectively randomized people without diabetes (n=26) with ACF to either treatment with metformin (250 mg/d, n=12) or no treatment (control, n=14).  The initial results are promising:

"At 1 month, the metformin group had a significant decrease in the mean number of ACF per patient (8.78 ± 6.45 before treatment versus 5.11 ± 4.99 at 1 month, P = 0.007), whereas the mean ACF number did not change significantly in the control group (7.23 ± 6.65 versus 7.56 ± 6.75, P = 0.609)."

In other words, this is the first reported trial showing that metformin can inhibit colorectal carcinogenesis in man. It also provides preliminary evidence that metformin suppresses colonic epithelial proliferation and rectal ACF formation and may be a potentially useful agent for early cancer chemoprevention.

Conclusions

In an accompanying editorial, Engelman and Cantley provided some useful commentary on the underlying pathways and highlighted the promise of metformin for cancer prevention and therapy in the lung and other sites.

Of course, no pharma company is going to sponsor large scale epidemiology trials as metformin is now available generically, but given the prominence given to chemoprevention by Harold Varmus in his NCI acceptance speech earlier this year, perhaps we will see some progress from both the NCI and the NIH in this field. It's really a public health issue that needs a broader perspective than individual companies can offer alone.

We all intuitively know that preventing or catching cancer as early as possible will likely yield better long term outcomes for patients than treating end-stage metastatic disease with highly expensive therapies.

 

Additional References:

ResearchBlogging.org Douglas JB, Silverman DT, Pollak MN, Tao Y, Soliman AS, & Stolzenberg-Solomon RZ (2010). Serum IGF-I, IGF-II, IGFBP-3, and IGF-I/IGFBP-3 Molar Ratio and Risk of Pancreatic Cancer in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology PMID: 20699371

Tao Y, Pinzi V, Bourhis J, & Deutsch E (2007). Mechanisms of disease: signaling of the insulin-like growth factor 1 receptor pathway–therapeutic perspectives in cancer. Nature clinical practice. Oncology, 4 (10), 591-602 PMID: 17898809

Chitnis MM, Yuen JS, Protheroe AS, Pollak M, & Macaulay VM (2008). The type 1 insulin-like growth factor receptor pathway. Clinical cancer research : an official journal of the American Association for Cancer Research, 14 (20), 6364-70 PMID: 18927274

Douglas JB, Silverman DT, Pollak MN, Tao Y, Soliman AS, & Stolzenberg-Solomon RZ (2010). Serum IGF-I, IGF-II, IGFBP-3, and IGF-I/IGFBP-3 Molar Ratio and Risk of Pancreatic Cancer in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology PMID: 20699371

1 Comment

"LKB1 is a master kinase"

What a great subheader in a paper last year by Reuben Shaw (journal link below).

Liver kinase B1 (LKB1) first got my attention at the AACR lung cancer meeting in San Diego earlier this year, when a couple of translational researchers mentioned it during informal discussions about how it might play a critical but subtle role in lung cancer and potentially other cancers.

Looking at the literature, LKB1 was first identified as a tumor suppressor gene on human chromosome 19p13, responsible for the inherited cancer disorder Peutz-Jeghers Syndrome (PJS).  However, the interest at the AACR meeting centred around it being one of the most commonly mutated genes in sporadic human lung cancer, including some subtypes of non-small cell lung carcinoma (NSCLC).

Of course, being very interested in potential druggable targets, I was trying to get my head around this particular kinase.  Several scientists and researchers explained to me patiently that LKB1 is involved in energy levels and metabolism, rather than cell signalling per se, so it kind of went by the wayside as other interesting targets came up lately, associated with small molecule tyrosine kinase inhibitors (TKIs) or monoclonal antibodies.

Still, the fact that LKB1 and AMPK control cell growth in response to environmental nutrient changes stuck in the back of my mind while I quietly wondered whether it would eventually have it's day.

Fast forward to an AACR press conference this morning about the role of metformin, a biguanide therapy for managing hyperglycemia and diabetes, in the role of chemoprevention.  I'm going to write more about that meaty topic in another more detailed post tomorrow, but what fasinated me was the mention by Dr Michael Pollak about metformin altering cell energy levels, ie a control system that senses cell energy supplies and low reserves.  

It was also mentioned that the activation of the LKB1-AMPK pathway downregulates gluconeogenesis.  This process represents the export of energy from hepatocytes to the organism in the form of glucose.  In turn, this reduces blood glucose concentration, which results in a secondary decrease in insulin level.  

Essentially, the inhibition of hepatic gluconeogenesis is now felt to be a key process underlying the utility of biguanides in the therapy of type II diabetes.

What is interesting on several levels is:

  • Studies showing raised levels of free or circulating IGF1 may be associated with an increased risk of developing cancer
  • Epidemiology studies amongst people with diabetes taking metformin who may have a lower risk of developing cancer

Of course, when we look at the broader picture, we can see the interactions across several pathways, which makes the whole situation highly complex:

Picture 9
Source: University of Dundee

Clearly, there is now enough evidence to warrant researching the effects of metformin in cancer prevention, especially given that it is orally available, has had no long term safety issues and is now generically available.  These factors, coupled with a greater understanding of the biology of the involved pathways may make a productive new area of cancer research.

Tomorrow, I will cover the latest research involving metformin for chemoprevention in colorectal and lung cancers in more detail.

 

ResearchBlogging.org Shaw RJ (2009). LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta physiologica (Oxford, England), 196 (1), 65-80 PMID: 19245654

Shackelford DB, & Shaw RJ (2009). The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nature reviews. Cancer, 9 (8), 563-75 PMID: 19629071

1 Comment

Last week there was lot of excitement and interest surrounding the blog post on Roche/Plexxikon's data on PLX4032 in metastatic melanoma published in the New England Journal of Medicine. A number of the discussions on Twitter and email centred around what is causing resistance to the BRAF inhibitor?

If we take a look at the BRAF pathway alone, we would get a sense of the flow from the PDGF ligand through RAS, RAF and MAPK, which essentially drives angiogenesis and proliferation, like this 2004 review article:

Picture 5
Source: Nature Reviews Cancer

However, what this sort of simple diagrammatic picture doesn't tell us though, is where cross-talk or feedback loops might interfere with the inhibition to enable cell signalling to continue, thereby guaranteeing the tumour's continued survival despite our efforts to shut it down.

Another way of looking at the problem is to consider how pathways related to RAS signalling might possibly interact with the process, like this:

RASSource: Reaction Biology

When we look at the cell signalling processes this way, we can see that inhibiting PI3-kinase or AKT at the same time as RAF might turn out to be a useful approach.

It was therefore with great interest while browsing my RSS cancer feeds that I spotted a paper by Shao and Aplin in the current Cancer Research journal entitled: "Akt3-Mediated Resistance to Apoptosis in B-RAF–Targeted Melanoma Cells" (see journal link below).

If we use a BRAF inhibitor such as PLX4032 in advanced melanoma, we are effectively trying to kill the cancer cells by inducing apoptosis, or programmed cell death, thereby reducing the tumour growth and proliferation.  However, as the researchers put it very succinctly:

"Melanoma cells are highly resistant to anoikis, a form of apoptosis induced in nonadherent/inappropriate adhesion conditions.  Depleting B-RAF or the prosurvival Bcl-2 family protein Mcl-1 renders mutant B-RAF melanoma cells susceptible to anoikis."

They therefore began looking at different approaches to dealing with anoikis using both inhibitors and RNA interference.

These concepts may help us better understand how mutant B-RAF protects melanoma cells from apoptosis, thereby providing insight into possible resistance mechanisms to B-RAF inhibitors by developing new mutations in AKT. It also paves the way for future therapeutic strategies, either in combination, or in sequence, with a V600E BRAF inhibitor such as PLX4032 and an Akt inhibitor. I'm also wondering what effect a PI3-kinase inhibitor might have, but clearly adding an Akt inhibitor to the mix may well be worth a try in the first instance.

There are a number of AKT inhibitors in development in various companies pipelines. This is where the challenges and hurdles begin if a company doesn't have a relevant inhibitor because traditional R&D focuses on developing one's own pipeline with or without the current standard of care rather than cross-development with other companies with novel combination treatments unless a partnership, particularly with a small biotech, is specifically sought out.

As far as I know, Roche/Genentech or Plexxikon don't have an AKT in their pipelines, but there are some currently in early clinical development with other companies. The PI3K-mTOR-AKT pathway has been discussed in more detail in previous blog posts.

The most interesting and promising compound in this class is probably Merck's MK-2206, currently in phase II development for a number of different tumour types.

Other Akt inhibitors in R&D include:

  • Keryx: perifosine in phase II development for myeloma and colorectal cancer
  • Rexahn: Archexin in phase II trials in pancreatic cancer
  • Nerium: oleandrin in phase I development for metastatic renal and colorectal cancers
  • GSK: GSK2141795 and GSK21110183 are both in phase I trials for either hematologic malignancies or solid tumours

GSK had an earlier Akt inhibitor in phase I, GSK690693, but I believe it may have been terminated due to problems with hyperglycaemia, a common problem associated with PI3K-IGF-1R feedback. This problem has since been addressed and managed in other trials associated with these agents by the simple addition of metformin in the protocol. GSK now appear to be focusing on the follow-on compounds instead.

All in all, it's interesting to see how our knowledge of the biochemical and molecular pathways helps us better understand how cancer works and how we can use the data to devise improved strategies for tackling melanoma in the future. I'll be watching how this field develops with close interest.

 

ResearchBlogging.org Dibb, N., Dilworth, S., & Mol, C. (2004). Opinion: Switching on kinases: oncogenic activation of BRAF and the PDGFR family Nature Reviews Cancer, 4 (9), 718-727 DOI: 10.1038/nrc1434

Shao, Y., & Aplin, A. (2010). Akt3-Mediated Resistance to Apoptosis in B-RAF-Targeted Melanoma Cells Cancer Research, 70 (16), 6670-6681 DOI: 10.1158/0008-5472.CAN-09-4471

Crouthamel, M., Kahana, J., Korenchuk, S., Zhang, S., Sundaresan, G., Eberwein, D., Brown, K., & Kumar, R. (2009). Mechanism and Management of AKT Inhibitor-Induced Hyperglycemia Clinical Cancer Research, 15 (1), 217-225 DOI: 10.1158/1078-0432.CCR-08-1253

2 Comments

“Here’s to the crazy ones. The misfits. The rebels. The troublemakers. The round pegs in the square holes. The ones who see things differently. . . . While some may see them as the crazy ones, we see genius. Because the people who are crazy enough to think they can change the world are the ones who do.”

via bits.blogs.nytimes.com

The crazy ones are the ones who make things happen and get things done… in business, in Pharmaland, in clinical research even.

Long live the crazy ones!

(Hat tip: @mona)

Interesting news arrived in my email box this morning:

"Roche (SIX: RO, ROG; OTCQX: RHHBY) today announced that the U.S. Food and Drug Administration (FDA) issued a Refuse to File letter for accelerated approval for the company’s trastuzumab-DM1 (T-DM1) Biologics License Application (BLA). As planned Roche will continue with its ongoing Phase III EMILIA registration study. Roche will continue to work with the FDA and expects a global regulatory submission of T-DM1 mid 2012.

The BLA submitted in July 2010 requested accelerated approval for T-DM1 based on the results of a single-arm Phase II study, which showed T-DM1 shrank tumors in one-third of women with advanced HER2 positive breast cancer, who had received on average seven prior medicines, including two HER2 targeted agents."

There was a lot of excitement around the initial phase II results (one third of the women experienced tumour shrinkage, for example).  Yet, to much surprise, the application was rejected:

"In their review of the BLA, FDA stated the T-DM1 trials did not meet the standard for accelerated approval because all available treatment choices approved for metastatic breast cancer, regardless of HER2 status, had not been exhausted in the study population."

At first reading of the press release, my thoughts centred around:

  1. How could the FDA possibly reject such an application?
  2. Was an SPA agreed up front?

The first question is easy to be indignant about, but what criteria were used for the trial and what does the data actually show?

Looking at the available clinical trials for T-DM1, this one looks most likely as 100 patients were required and the inclusion criteria stated:

  • HER2-positive disease
  • Metastatic breast cancer
  • Disease progression on the last chemotherapy regimen received in the metastatic setting
  • Prior treatment with an anthracycline, trastuzumab, a taxane, lapatinib, and capecitabine in the neoadjuvant, adjuvant, locally advanced, or metastatic setting and prior treatment with at least two lines of therapy (a line of therapy can be a combination of two agents or single-agent chemotherapy) in the metastatic setting
  • At least two lines of anti-HER2 therapy must have been given in the metastatic setting as monotherapy or combined with chemotherapy or hormonal therapy. The HER2-targeted agent can include trastuzumab, lapatinib, or an investigational agent with HER2-inhibitory activity.

There are only two approved treatments for HER2-positive breast cancer, trastuzumab and lapatinib, plus others in clinical trials, eg pertuzumab, which was also allowed as one of the prior therapies.  All patients appear to have been refractory to at least two of these drugs, most likely trastuzumab and lapatinib.

The prior chemotherapies included anthracyclines, taxanes and capecitabine, which is quite heavy pre-treatment and includes all of the considered standards of care for several lines of therapy.  Indeed, the results of the trial presented at the San Antonio Breast Cancer Symposium last December showed that the average number of prior treatments in the metastatic setting was 7.

The others that could be used in the treatment of breast cancer include nab-paclitaxel (Abraxane), which I'm assuming would be covered in the taxane group and ixabepilone (Ixempra), an epothilone approved in by the FDA for metastatic breast cancer following progression on a taxane and anthracyclines, with or without capecitabine.

Ixabepilone is not a taxane or an anthracycline and therefore technically not covered in the inclusion criteria as a prior therapy, although some of the women would likely have received it, but not all.  It is, however, a taxane-like compound in that it targets the microtubules, as described in the PI:

"Ixabepilone is a semi-synthetic analog of epothilone B. Ixabepilone binds directly to B-tubulin subunits on microtubules, leading to suppression of microtubule dynamics. Ixabepilone suppresses the dynamic instability of aB-II and aB-III microtubules."

If that was the criteria for rejecting the trial as not truly refractory in a very heavily pre-treated setting, I would be surprised.  It's a bit of a technicality and nit-picking. Ixabepilone is not a commonly preferred treatment at all and BMS reported very low sales in 2009.  Most US physicians appear to prefer the well established taxanes such as paclitaxel, docetaxel and nab-paclitaxel rather than a synthetic taxane-like agent and probably consider it as a last resort. It is used as a standalone therapy in the refractory/salvage setting.

However, we're talking about a potential indication for a targeted agent that inhibits HER2 dimerization not a chemotherapy, so I would have thought that most of the important bases were covered by including the most common chemotherapies, trastuzumab and lapatinib in the inclusion criteria.

With regards to the latter question, Twitter chat this morning suggested that no, an SPA wasn't formally agreed upon, but Roche held discussions with the FDA that led them to file for accelerated approval. Thanks to Adam Feuerstein of The Street for being the first to answer that question.

My general opinion is that if you have an agreed Special Protocol Assessment (SPA) and meet the defined targets, it's much easier to move forward and gain approval. If you don't, things may turn into a crapshoot and it can go either way.  And that seems to be what has happened here.

The other factor that comes into play in this discussion is the ongoing discussion of the accelerated approval for Avastin in metastatic breast cancer and the overwhelming negative ODAC opinion last month.  That will not have helped, although one would like to think it shouldn't influence any decision making at the FDA.  We would probably be naive to think otherwise given the full protocol included PFS rather than OS as the endpoint.

To be fair, Roche appear to be addressing this issue, in their announcement this morning:

"Roche will amend the Phase III randomized EMILIA study in order to rigorously evaluate overall survival in addition to progression-free survival and will submit data from EMILIA to support a global regulatory submission in mid 2012."

Overall, I think it's a disappointing decision by the FDA given the heavily pre-treated population and lack of options for women who are refractory to both trastuzumab and lapatinib. Chemotherapy has not been shown to be particularly effective in HER2 disease, and by then the women generally have a poor prognosis. When you look again at the SABCS data, you find an objective response rate (ORR) of approximately 30% and a clinical benefit rate of around 40%, which is quite startling in a heavily pre-treated group.

Meanwhile, for now we'll have to wait another two years to see what the survival data looks like.  That's a rather long time for women who have failed Herceptin and Tykerb to wait for a drug that appears to have significant activity.

{UPDATE: I posted this analysis in a hurry, uncaffeinated, and in a rush to head out to a meeting.  Of course, one remembers later that SPA's apply to phase III not phase II trials, so a formal agreement wouldn't apply here. Thanks also to the two people who gently reminded me of this. Usually though, there are some discussions with FDA around the trial design for accelerated review. Essentially, in layman terms, it means patients are refractory to existing treatments for the disease concerned. Note that Roche, in their press release defined it as, "Consideration by the FDA for accelerated approval requires recognition of a defined patient population of unmet need (a life-threatening disease with limited treatment choices)." It looks like the FDA are applying it more strictly to ALL therapies, although the number of women who might have taken ixabepilone AND be HER2-positive will likely be miniscule.  I would still maintain that refractory to all available HER2 therapies and most chemotherapies apply for the majority in this case.}

3 Comments

Treatment for ovarian cancer hasn't changed much in the last ten years, reflecting the lack of biomarkers and biochemical targets for the disease. Chemotherapy with a platinum (carboplatin or cisplatin) and a taxane (paclitaxel or docetaxel) has therefore formed the bedrock of therapy, along with other options such as gemcitabine or pemetrexed, as illustrated in the latest NCCN Guidelines.

The good news is that the use of paclitaxel-based combination chemotherapy has been shown to increase progression free survival (PFS) and overall survival (OS) in women with primary peritoneal or ovarian cancers.

While a proportion of ovarian cancers have been shown to be highly chemosensitive, a large number unfortunately fail to respond to primary taxane therapy, leading to the emergence of resistant disease.

The big unanswered questions are therefore why does this happen and what can be done about it to improve outcomes and overall prognosis?

It was with great interest that I read about the findings of a new study just published in Cancer Cell from researchers at MD Anderson (see e-link in the references below).  According to the MD Anderson news alert:

"For the first time, Salt Inducible Kinase 2 (SIK2) has been found to play a critical role in cell division and to regulate the response of some ovarian cancers to chemotherapy."

It's not often when you see the mention of both a potential target and prognostic/predictive biomarker mentioned in the same sentence as ovarian cancer, so this is huge news!  The press release went on to claim:

"Researchers found that depleting SIK2 from ovarian cancers sensitized the cancer cells to paclitaxel, a commonly prescribed chemotherapeutic agent that inhibits cell division, making the drug more effective in stopping the cancer's growth. Levels of the SIK2 protein are increased in approximately 30 percent of ovarian cancers and are associated with poorer survival in women with the disease."

The researchers analysed nearly 780 pools of siRNAs to identify proteins that alter sensitivity to paclitaxel. They found that SIK2 regulates sensitivity to paclitaxel and prevents cell division. This means that SIK2 may offer a useful therapeutic target for pipeline drugs to be developed in ovarian cancer.

What was even more fascinating was that another related article on ovarian cancer from Bast's group appeared in the same journal. In essence, they used siRNA-loaded nanoparticles to stifle a protein, Zeste homolog 2 (EZH2), which is associated with poor survival. This resulted in inhibition of angiogenesis (formation of new blood vessels) to the tumour and caused a steep reduction in the tumour burden in a mouse model of ovarian cancer.

In this study, the authors looked at human 180 ovarian cancer tumours and found that the protein was overexpressed in the tumour samples (66%) and in the endothelial cells (67%). It is relevant to note that endothelial cells line the inside of blood vessels and play a crucial role in angiogenesis.

In practice, they found that women with increased EZH2 levels in their tumours had a median survival of 2.5 years compared to 7.33 years for those without. Looking at overexpression in the endothelial cells, the difference was 2.33 years versus 8.33 years for those with normal levels.

Like me, you're probably wondering how these nanoparticles work.  According to MD Anderson:

"The nanoparticles accumulate in the cancer cell and vasculature passively as they circulate in the blood stream. Chitosan nanoparticles are so small that they can flow through tiny holes in the tumor vasculature. They also accumulate in other organs, so the researchers are working to add a targeting molecule that will limit nanoparticle uptake to tumors and their vasculature."

Targeting EZH2 may have application beyond ovarian cancer, since it been associated with the progression and spread of bladder, breast, prostate and gastric cancers and cancer of the pharynx.

All in all, a really interesting pair of papers from Bast's group, which may have clinical promise and real application to the future treatment of ovarian cancer.

 

ResearchBlogging.org Ahmed, A., Lu, Z., Jennings, N., Etemadmoghadam, D., Capalbo, L., Jacamo, R., Barbosa-Morais, N., Le, X., Vivas-Mejia, P., & Lopez-Berestein, G. (2010). SIK2 Is a Centrosome Kinase Required for Bipolar Mitotic Spindle Formation that Provides a Potential Target for Therapy in Ovarian Cancer Cancer Cell, 18 (2), 109-121 DOI: 10.1016/j.ccr.2010.06.018

Lu, C., Han, H., Mangala, L., Ali-Fehmi, R., Newton, C., Ozbun, L., Armaiz-Pena, G., Hu, W., Stone, R., & Munkarah, A. (2010). Regulation of Tumor Angiogenesis by EZH2 Cancer Cell, 18 (2), 185-197 DOI: 10.1016/j.ccr.2010.06.016

1 Comment

Hot on the heels of last week's New England Journal of Medicine article on ipilimumab (BMS) comes another article on metastatic melanoma, this time from Keith Flaherty's group in Pennsylvania and Boston on BRAF inhibition with PLX4032, an exciting compound being developed by Plexxikon/Roche (see link below in the references for the article).

I've written a few posts on this interesting compound recently (e.g. here and here), for those interested in getting up to speed on the concept and data.

The basic concept is that a while back it was noticed that some tumours such as melanoma develop the V600E BRAF mutation and induce resistance or a poorer prognosis, so common sense says why not find a targeted agent to inhibit the activity to see what happened?

In the August 26th edition of the NEJM, Flaherty et al., described the updated phase I results in two phases:

  • 55 patients (49 with melanoma) in the dose escalation phase
  • 32 additional people with metastatic melanoma who had BRAF with the V600E mutation in the extension phase

Now, remember, a phase I trial usually seeks to define the maximum tolerated dose (MTD), which is used in the phase II studies and looks at the general tolerability and side effects.  Efficacy is not the primary end point, but of course, we all secretly look at the activity to see if there is any sign of a response!

First things first.  After some palaver with the crystalline formulation, it was finally settled on a "micro-precipitated bulk powder" since that offered superior bioavailability. I'm paraphrasing a bit and taking liberties here, but no doubt regular readers will sense my excitement and tendency to skip to the bottom line and find out what it all means, although we have all been there with those kind of challenging problems, especially as they add delays and frustrations all around!

Anyway, as a result of these changes, the final recommended dose that emerged for the phase II studies was 960mg BID, with increases in the dose limited by grade 2 or 3 rash, fatigue or arthralgia. These kind of side effects are fairly typical of oral TKI's and very different from what we saw with last week's NEJM article on immunotherapy with ipilimumab in a similar population.

Potentially, the biggest concern is the appearance of squamous-cell carcinoma that appeared in 10 of 32 patients (31%) in the dose escalation cohort.  While normally well differentiated and of low invasive potential, it is something to note.  The authors noted that recent data earlier this year (see references below) suggest BRAF inhibitors can activate the MAP kinase pathway in cells that lack a BRAF mutation and may explain some of the peculiar side effects seen with PLX4032.

Previously, Flaherty et al., reported nine responses in the second PLX4032 cohort and median PFS of 6 months.  For comparison with the updated data, see the last report before reading on. Over half of the people in both cohorts had received 2 or more therapies, so these are a mix of relapsed and refractory patients:

  • In the first cohort of 55 people, there were 16 with the BRAF V600E mutated melanoma and received 240mg or more of PLX-4032.  The efficacy?  Well, there were 10 partial response and 1 complete response (69%).
  • In the extension cohort of 32 people, 24 were partial responders and 2 had a complete response (81%), which is pretty impressive all around.  No wonder Dr Flaherty was very excited when interviewed for an article in the NY Times earlier this year!

My first reaction was slight disbelief, after all, this is a very difficult to treat and highly aggressive disease, thus sadly, people do tend to relapse early. In short, if there were three cancers I absolutely wouldn't want to get, this would be one of them.

On closely checking the data carefully including the 81% response rate in the second cohort (yes, it's correct!), I noticed that the researchers reported across all patients in the phase I study, the median progression free survival (PFS) was now improved to 7 months.  That means that 50% did worse and 50% did better than 7 months, way better than one might expect so early in a trial.  I did double check again and pinch myself, as it was late on Tuesday night when I penned the draft.

If you are interested in this area, do check out the link to the article below because the water plots and anti-tumour responses over time are well worth looking at, especially as some people are clearly achieving responses approaching a year, despite having advanced disease.

"Responses were observed at all sites of the disease, including the bone, liver, and small bowel."

Yes, it is still very early, but how awesome is it to read that?

As an aside, a number of readers have written asking why sorafenib hasn't shown to be effective in melanoma, despite inhibiting BRAF.  Flaherty et al., had an answer for that. They suggested that  because it also inhibits other pathways, it may well be that the non-BRAF effects of the drug mediate side effects that limit the ability to achieve enough drug concentration and thus the drug concentration isn't high enough to effectively inhibit the V600E BRAF mutation.  An interesting theory that also speak to the idea that several specific inhibits may be more effective that more promiscuous multi-kinase inhibitors.

The good news is that for now at least, we seem to be on the right track with PLX4032 and ipilimumab in metastatic melanoma.  It will be interesting to see what the mechanisms of resistance are down the road, and whether we have some options in the works for either combination or sequencing of different targeted agents for this disease.

 

ResearchBlogging.org Flaherty, K., Puzanov, I., Kim, K., Ribas, A., McArthur, G., Sosman, J., O'Dwyer, P., Lee, R., Grippo, J., Nolop, K., & Chapman, P. (2010). Inhibition of Mutated, Activated BRAF in Metastatic Melanoma New England Journal of Medicine, 363 (9), 809-819 DOI: 10.1056/NEJMoa1002011

Heidorn, S., Milagre, C., Whittaker, S., Nourry, A., Niculescu-Duvas, I., Dhomen, N., Hussain, J., Reis-Filho, J., Springer, C., & Pritchard, C. (2010). Kinase-Dead BRAF and Oncogenic RAS Cooperate to Drive Tumor Progression through CRAF Cell, 140 (2), 209-221 DOI: 10.1016/j.cell.2009.12.040

Poulikakos PI, Zhang C, Bollag G, Shokat KM, & Rosen N (2010). RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature, 464 (7287), 427-30 PMID: 20179705

Hatzivassiliou, G., Song, K., Yen, I., Brandhuber, B., Anderson, D., Alvarado, R., Ludlam, M., Stokoe, D., Gloor, S., Vigers, G., Morales, T., Aliagas, I., Liu, B., Sideris, S., Hoeflich, K., Jaiswal, B., Seshagiri, S., Koeppen, H., Belvin, M., Friedman, L., & Malek, S. (2010). RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth Nature, 464 (7287), 431-435 DOI: 10.1038/nature08833

2 Comments

At the annual American Association of Cancer Research (AACR) annual meeting earlier this year, Prof Bert Vogelstein presented a fascinating lecture on the critical cancer pathways and how targeting the aberrant signalling may potentially lead to new breakthroughs in treatment.  I've been meaning to write a series on those particular pathways, but things have been very busy since the conference in DC!

It was therefore with great interest that a new paper came out yesterday in the Cancer Research journal entitled, "Gamma-Secretase Inhibitors Enhance Temozolomide Treatment of Human Gliomas by Inhibiting Neurosphere Repopulation and Xenograft Recurrence."

This is also hot on the heels of negative news from Lilly the other week regarding their gamma secretase inhibitor, semagacestat, in Alzheimer's disease. Now, if I were on their oncology team, I'd snap it up and investigate the possibilities in cancer indications, because one's man's poison is another man's nectar: there aren't too many NOTCH inhibitors in oncology development that I know of, and those that are, are still in relatively early phase I development.  More about the pipeline compounds later.

So what's all the fuss about and how does γ-Secretase connect with NOTCH signalling?  Let's take a look at the basic pathway:

Picture 1Source: Cell Signal

NOTCH signalling is an evolutionary pathway that has been shown to regulate cell-fate determination (renewal) during development and also in stem cells.  Without going into too much biochemistry, it essentially enables cell-cell communication and continually enables renewal of adult tissues such as blood, skin, and gut epithelium not only to maintain stem cells in a proliferative, pluripotent, and undifferentiated state.

You can gather from this, therefore, that aberrant NOTCH signalling might also drive or be involved with the dreaded word in cancer: proliferation.  Why?  Because Notch signaling appears to play a role in regulating the cellular actions of VEGF, ie angiogenesis. For those interested in this area, there is a link to a review article on angiogenesis and NOTCH below.

You can also see from the picture above that gamma secretase is a protease that cleaves the NOTCH ligand across the cell membrane.  How then does this relate to gliomas?  According to the study authors:

"Notch activity is upregulated in many gliomas and can be suppressed using gamma-secretase inhibitors (GSI)."

What they found was very interesting:

Basically, in a mouse xenograft model adding a gamma-secretase inhibitor to a standard glioma drug, temozolomide, reduced tumour growth and recurrence and increased survival more effectively than either drug alone.  When you consider that NOTCH may play a role in angiogenesis, these findings make a lot of sense.

What about NOTCH inhibitors in the pipeline?

Merck (MK0752)

This is good news for Merck in particular, since they market temozolomide, a standard treatment for gliomas and have a gamma-secretase inhibitor, MK-0752, in phase I development for breast and pancreatic cancers.  There is also a single agent phase I dose finding trial ongoing with recurrent or refractory CNS tumours, but the new data from Cancer Research may excite their scientists to consider combining MK-0752 with temozolomide in gliomas.  It's certainly worth a shot.

Lilly (semagacestat)

We mentioned Lilly's agent, semagacestat, but as far as I know that is only being tested in Alzheimer's disease although they appear to be testing a NOTCH inhibitor in oncology, as this advanced solid tumour trial suggests.  It may a different compound, however, as the agent is not named.

Roche/Genentech (RO4929097)

The only other NOTCH inhibitor I'm aware of is from Roche/Genentech (RO4929097), which is being tested in a much broader range of cancers than Merck's, including a trial about to start in newly diagnosed gliomas, with temozolomide and a phase II study as a single agent in relapsed/refractory glioblastomas.  Nice work, Roche/Genentech!

If you know of any other NOTCH or gamma secretase inhibitors in development for cancer indications, do drop a note in the comments.  I'm sure Vogelstein would agree that this is a very promising area of research indeed so far.

 

ResearchBlogging.org Gilbert, C., Daou, M., Moser, R., & Ross, A. (2010).  -Secretase Inhibitors Enhance Temozolomide Treatment of Human Gliomas by Inhibiting Neurosphere Repopulation and Xenograft Recurrence Cancer Research DOI: 10.1158/0008-5472.CAN-10-1378

Cook, K., & Figg, W. (2010). Angiogenesis Inhibitors: Current Strategies and Future Prospects CA: A Cancer Journal for Clinicians, 60 (4), 222-243 DOI: 10.3322/caac.20075

4 Comments

Regular readers will know that I'm a big fan of Gaping Void cartoons by Hugh Macleod.

This one really resonated:

Picture 1

Source: Hugh Macleod, Gaping Void

If you think about it, Pharma R&D is also a bit of an adventure (or even a roller coaster sometimes); you never know quite whether you're on a winner or a loser with company pipelines and portfolios.

Attitude also comes into it – do you live by paycheck or project, or do you truly love what you do?

 

Nanotechnology is a topic that fascinates me, ever since hearing some interesting data in pancreatic cancer at the American Association of Cancer Research (AACR) meeting last November on Molecular Targets in Boston.

Someone kindly sent me a paper from PNAS recently (link below), from 2008, which got my attention essentially because:

"Integrin aBv3 is found on a subset of tumor blood vessels where it is associated with angiogenesis and malignant tumor growth. We designed a targeted aBv3 nanoparticle (NP) encapsulating the cytotoxic drug doxorubicin (Dox) for targeted drug delivery to the expressing tumor vasculature."

Now, giving doxorubicin by the normal route of administration results in noticeable weight loss and other side effects when given systemically.  Using nanotechnology, a 15-fold increase in anti-metastatic activity without concomitant weight loss, which is very interesting indeed.

The authors noted that integrin could represent an ideal vascular targeting receptor since it is highly expressed on the angiogenic endothelium and expression of this receptor on tumor vessels correlates with disease progression.  This was first shown by Gasparini et al., in 1998 (see reference source below).

Meanwhile, in this 2008 study, the researchers used models from renal cell and pancreatic carcinoma to determine the effectiveness of the nanoparticle delivery system. Given that the results appear better with metastatic rather than primary tumour growth, I wondered if anyone was looking at this concept with currently approved drugs for metastatic disease?

A quick search for nanoparticle and cancer in the clinical trials database brought up mostly trials associated with nanoparticle paclitaxel (Abraxane) from Abraxis ($ABII). Of course, one of the challenges here is that in animal research, drugs and nanoparticles can be injected directly into the tumour thus avoiding systemic effects, whereas in human research, nanoparticle drugs are more typically combined with regular drugs and infused as normal.

It was, hover, no surprise to learn earlier this year that Abraxane is looking promising in 2nd line advanced pancreatic cancer as a single agent after failure of gemcitabine-based chemotherapy.  At AACR last year, researchers from Mass General showed that Abraxane targets the tumour stroma, depleting it, thereby reducing the impediment to chemotherapy.  The Nab-paclitaxel utilises endogenous albumin pathways via binding of the albumin to secreted proetin acid rich in cysteine or SPARC.  Interestingly, pancreatic cancer is known to overexpress SPARC and therefore offers a logical target for the drug.  Abraxis reported in May that the median overall survival was 7.3 months, which is impressive in a very hard to treat refractory population.

I wonder what the Nab paclitaxel data would look like in front-line treatment in combination with gemcitabine?  Or what about using it in advanced renal cancer with any of the half dozen drugs already approved but with limited effectiveness? Would adding a nanoparticle delivery system such as Abraxane to the mix reduce metastasis, deplete the stroma and improve access to the tumour for the anti-angiogenic agents?

Overall, it's an interesting technology concept for improving drug delivery and we're still at relatively early stages of research.  Hopefully, we'll hear more about nanotechnology and the broader application of new combination nanoparticles to cancer treatment in the near future.  It takes significant investment, creativity in trial design and an intensive yet broad research program to drive results though. Celgene ($CELG) have since bought Abraxis and may be more willing to aggressively invest in the growth of this interesting drug.

I must say that the AACR Molecular Targets and Cancer Therapeutics meeting every November is one of my all time favourites, but it alternates with Europe every other year and will be in Berlin this November, so depending upon my schedule I may not be able to make it this time 🙁

 

ResearchBlogging.org Murphy, E., Majeti, B., Barnes, L., Makale, M., Weis, S., Lutu-Fuga, K., Wrasidlo, W., & Cheresh, D. (2008). From the Cover: Nanoparticle-mediated drug delivery to tumor vasculature suppresses metastasis Proceedings of the National Academy of Sciences, 105 (27), 9343-9348 DOI: 10.1073/pnas.0803728105

Gasparini G, Brooks PC, Biganzoli E, Vermeulen PB, Bonoldi E, Dirix LY, Ranieri G, Miceli R, & Cheresh DA (1998). Vascular integrin alpha(v)beta3: a new prognostic indicator in breast cancer. Clinical cancer research : an official journal of the American Association for Cancer Research, 4 (11), 2625-34 PMID: 9829725

error: Content is protected !!