Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts tagged ‘PI3-kinase’

Here’s a quick update on the next conference I’m planning to attend in New York next week.  It’s hosted by the New York Academy of Sciences (NYAS) in their downtown New York headquarters by the World Trade Center, which has fantastic panoramic views of uptown Manhattan and Brooklyn from the 40th floor.  More importantantly though, judging by the last few meetings I’ve attended there on cancer metabolism and a most fascinating lecture on ink and tattoos from Carl Zimmer, it should be a very good event and well worth attending.

The latest conference is a two day affair on “Inositol Phospholipid Signaling in Physiology and Disease” otherwise known as the PI3K-AKT-mTOR pathway, which is a key process that is dysregulated in many cancers:

PI3K mTOR AKT Cancer Signaling Pathway Conference

The organizing committee of William Kerr (Suny), Christina Mitchell (Monash Univ) and Christian Rommel (Intellikine) have done an excellent job putting together a comprehensive program that covers a wide variety of related topics from both academia and industry across the globe.

I’m really looking forward not only to the science feast, but also to the networking opportunities to mix and mingle with some of the top researchers in the PI3K field, including Lew Cantley (Harvard), Neal Rosen (MSKCC), Bart Vanhaessebroeck (Barts, London) and David Solit (MSKCC) amongst many others.

For those of you interested in registering for this event, you can obtain a 15% discount when you click on the graphic or link above and enter the coupon code INOSITOL15 on checkout, as I’m delighted to say Icarus Consultants was invited to be one of the media partners for the event.

For those who cannot attend, I’ll post a short synopsis of the conference on PSB after the event later next week.  In the meantime, I hope to see you there!

While thoughts have already turned to the forthcoming ASCO 2012 meeting and today I am off to AUA 2012 in Atlanta, the annual meeting of the American Association Association for Cancer Research (AACR) last month continues to generate insights.

At AACR I was delighted to meet up with Philippe Aftimos, MD, a Clinical Research Fellow at the Jules Bordet Institute in Brussels, Belgium.   Philippe is medical coordinator of the Clinical Research Unit and someone who I met through social media (@aftimosp), so it was a pleasure to meet in person.

I was, therefore, thrilled when Philippe agreed to do a guest blog post about what he thought were the highlights of AACR 2012:

 

A Medical Oncologist at AACR 2012 in Chicago by Philippe Aftimos, MD

As a medical oncologist with recent interest in clinical research, I attended the AACR annual meeting for the first time in Chicago from March 31st to April 4th 2012.

Scientists on the road to end cancerAt first view, I was impressed by the enormous organization and very large number of participants, which was at least as important as the ASCO annual meeting. However, as a clinician, I only recognized very few familiar faces as the large majority of attendance included basic and translational scientists, as well as representatives of pharma.

Simultaneous sessions took place all over McCormick place and were featured: daily plenary sessions, major symposia, minisymposia, forums, educational sessions, methods workshops, poster sessions, “Current Concepts and Controversies in Organ Site Research” sessions and “Current Concepts and Controversies in Diagnostics, Therapeutics, and Prevention series” sessions. Planning the day’s schedule and navigating between the different sessions was made easy by the well-designed AACR 2012 Annual Meeting app.

My highlights from AACR 2012 can be divided into 3 subjects:

1. Pathways and new drug development

  • The PI3K-AKT-mTOR pathway was the subject of multiple sessions. It is mutated in tumors such as breast, lung, ovarian, endometrial carcinomas as well as gliomas. Inhibition of TORC1 has been shown to release inhibition of PI3K. Cancer cells harboring low BIM, a pro-apoptotic Bcl-2 family member, are resistant to PI3K inhibitors. PI3K inhibitors can reverse resistance to anti-EGFR tyrosine kinase inhibitors (TKIs) and the combination with MEK inhibitors is active against K-Ras mutant NSCLC. Selective inhibitors of PI3K isoforms are currently in clinical development. BYL-719 is PI3K alpha inhibitor and has shown tumor shrinkage in the phase 1 setting. It is potent against mutated cells and has anti-angiogenic properties. HER-2 amplification and PIK3CA mutation predict sensitivity while PTEN, B-Raf and K-Ras mutations confer resistance. GSK-2636771 is a selective PI3K beta inhibitor potentially inducing synthetic lethality in PTEN deficient mice. Hyperglycemia and hyperinsulinemia are class adverse events of PI3K inhibitors but are seldom seen with GSK-2636771.
  • ABT-199 is a Bcl-2 inhibitor in phase 1 development with very promising activity starting from the first cohort (starting dose) of chronic lymphocytic leukemia patients. It has also shown synergy when combined with rituximab or combination chemotherapy. Activity is correlated to Bcl-2 expression and specific targeting of Bcl-2 reduces the incidence of thrombocytopenia.
  • With the discovery of new chemotherapy agents in the treatment of castrate-resistant prostate cancer, interest in anti-hormonal treatments has been renewed. AZD-3514 is a first in class selective androgen receptor downregulator (SARD). It targets the androgen receptor in the nucleus and is currently in phase 1 development.
  • PD-0332991 is a highly selective inhibitor of CDK4/CDK6 resulting in potent G1 arrest, especially in Rb positive tumors. It showed activity in phase 1 trials with stable disease in breast cancer and liposarcoma. The most impressive results were in the treatment of mantle cell lymphoma with 1 CR in one patient still on-study for 2 years, 2 PR with one patient on study for 30 months and 7 SD out of 16 patients. Main DLT was neutropenia. Thrombocytopenia was also a limiting factor.

2. Immune therapy

AACR 2012 Annual MeetingThe goal is to increase the tail of the curve in the photograph in the right. The approval of ipilimumab in the treatment of metastatic melanoma has inaugurated the new era of anti-cancer immune therapies.

They were very much put in the spotlight at AACR 2012 with a plenary session entitled: Immune Therapies: The Future Is Now. Highlights included: adoptive T-cell therapy, recombinant pox-viral vaccines, intra-tumor injection of vaccines, combination of vaccines and targeted agents in the treatment of melanoma. Characteristics of therapeutic vaccines are: minimal toxicity, indirect effects on tumors, delayed responses that increase over time. Administration in the early course of disease may be better and overall survival is usually the endpoint of clinical trials.

Agents that most caught my attention were monoclonal antibodies targeting the Programmed Death-1 (PD-1) T cell co-receptor and its ligand, B7-H1/PD-L1. Durable responses have been seen, often long-lasting off-therapy. Overall response rates as high as 30% have been demonstrated in renal cell carcinoma and melanoma. Tumor shrinkage was also seen in non-immunogenic cancers such as non-small cell lung cancer. Strong endogenous anti-tumor immune response upregulates PD-L1. For week endogenous anti-tumor immune response, combinations with epigenetic therapies may be key. Agents such as azacytidine can create an inflammatory response. Search for biomarkers such as PD-L1 expression and tumor-infiltrating lymphocytes is ongoing. This reference further explains this pathway: Topalian SL, Drake CG, Pardoll DM. Curr Opin Immunol. 2012 Apr;24(2):207-12.

3. Challenges for the future

With many clinical trials yielding negative results, the necessity for “smarter” trials has become evident. Trials of the future should be biomarker-stratified, enriched and adaptive. Histology-independent, aberration-specific trials should also be considered while being aware of different degrees of functionality and sensitivity of mutations. Combinations of targeted agents are the backbone of recent clinical trials and scientific rationle should be strong:

  • Supportive and confirmed in vivo and preclinical data
  • Suitable animal toxicity
  • Predictable biomarkers
  • Delivery to patients: number of pills, …

2 challenges remain:

  • Cooperation between different pharmaceutical patients developing the drugs of the combination
  • Regulatory-acceptance of mutation-based trials as well as regulations for targeted agents combinations

The session tackling the last issue deserved to feature as a plenary session. A very large crowd attended it with many participants finally sitting on the floor or leaning on walls.

Finally, the 2012 AACR Annual Meeting was yet another example of the rapid development and influence of social media with many scientists, clinicians and pharma representatives tweeting the information live, sharing their opinions, and interacting together.

Bio: Philippe Aftimos, MD

Medical Oncologist, graduate of Saint Joseph University in Beirut, Lebanon and the Free University of Brussels, Belgium. Trained: MD Anderson Cancer Center (Houston, Texas), Memorial Sloan-Kettering Cancer Center (New York City, New York) and Institut Gustave Roussy (Paris, France).

Currently, medical coordinator of the Clinical Research Unit at Institut Jules Bordet Cancer Centre in Brussels, Belgium, investigator for multiple clinical trials. My main interests are new drug development, phase 1 trials and breast cancer.

 

1 Comment

“I’ve missed more than 9000 shots in my career.  I’ve lost almost 300 games.  26 times, I’ve been trusted to take the game winning shot and missed.  I’ve failed over and over and over again in my life.  And that is why I succeed.”

Michael Jordan, Chicago Bulls

Michael Jordan, Chicago Bulls

Source: wikipedia

Continuing the sporting metaphors this week, I was catching up on blog reading last night and noticed that Jim Lefevere put up a nice post on Digital Strategist about how:

Domain Expertise + Work Ethic + Time = Success

He used Michael Jordan as an example to illustrate the competitiveness that is required for the top level.

While talking to scientists and researchers at the recent AACR PI3K-mTOR meeting about their myriad of iterative experiments with GWAS, Western Blots and such, you can imagine the parallels with scientific research.

It struck me how the scientists in this particular field of cancer research are both highly collaborative and competitive at the same time, while also being very focused and intense on the end game (implications for clinical research), perhaps more so than other sub specialty areas I’ve come across lately.

The main message I learned from the meeting can be summed up in this little forumla:

Driver Mutation + Adaptive Pathway + Ligand + Patient Selection = Possible Success

A lot of data on PI3K and mTOR can be expected at forthcoming annual meetings at AACR (April) and ASCO (June), so it will be interesting to see how the new combinations of PI3K or mTOR with AKT or MEK, for example, are panning out and in which tumour types.

 

 

1 Comment

Today, I’m heading off to San Francisco for the AACR Special Conference on Targeting PI3-Kinase and mTOR in cancer.  For those of you needing a brief primer on the pathway, you can find more about it in this 2010 post, which vies with one about ipilimumab in melanoma as the top two posts on Pharma Strategy since the end of October.

You can view the PI3K-mTOR program here.

I’m really excited to be attending this event – a lot of the ‘big guns’ in the PI3-kinase field are speaking at this event, including Lewis Cantley, Jeffrey Engelman, David Sabatini, Carlos Arteaga, Neal Rosen, Gordon Mills and many others.

There are also presentations from scientists at various Pharma and Biotech companies with PI3-kinase inhibitors in development, so it won’t just be about the basic translational research per se, but also about how the R&D is progressing to date with new therapeutics.

If anyone is at the meeting, please do stop and say hello – it’s always nice to meet readers in person – I bumped into a few at last weeks ASCO/ASTRO/SUO GU cancers symposium, for example.

I’ll be tweeting a few snippets from the conference, including tonight’s keynote by Jose Baselga (Mass General Hospital), but excluding unpublished data, under the hashtag #PI3K.  The aggregated tweets from that hashtag will be captured between now and Saturday in the widget below for easy following for those remote and interested in this sub-specialty:

1 Comment

Over the last couple of years, our knowledge and understanding of non-small cell lung cancer (NSCLC) has improved as mutations and translocations that drive tumour growth and survival have been identified.

Unfortunately, while we have many new targeted agents in the clinic, few have so far made it to market for broader use in every day clinical practice.  EGFR inhibitors such as erlotinib (Tarceva) and gefitinib (Iressa) were probably the first to gain people’s attention and soon we will hopefully have crizotinib for ALK translocations, since Pfizer have begun the rolling NDA submission to the FDA.

Lowly and Carbonne (2011) discussed the progress with lung cancer subsets in a short piece in Nature Reviews Clinical Oncology that is well worth checking out (see reference below).   They point out that identifying these groups based on their molecular peculiarities is important because patients can be identified and better response rates obtained in a more targeted population:

“Patients with lung cancers harboring EGFR mutations have dramatically greater clinical responses when treated with the oral EGFR tyrosine kinase inhibitors (TKIs) erlotinib or gefitinib, compared with patients without these mutations. In 2009, two seminal trials (the Iressa Pan-Asia Study [IPASS] and the Spanish Lung Cancer Group) demonstrated response rates of around 70% to EGFR TKIs in this cohort of patients, compared to a response rate of 30–40% with traditional platinum-based chemotherapy.”

They went onto describe the molecular subsets found to date:

Molecular Subsets in Lung Cancer

Note that approx. half of the aberrant mutations have yet to be found and about one-eighth have been identified, but clinical trials are still ongoing with various inhibitors, so there is more hope for the future if any of these pan out with positive data.

Of course, what everyone wants to know is what is the next target that may emerge after crizotinib and ALK.  I think PI3-kinase inhibitors look the most promising and there are a few being evaluated in trials right now.  However, my suspicion is that we will be very lucky to get it right first time and it may well take some more creative combinations than at present before we figure it out.  We may see a few failures before someone cracks the optimal solution based on biomarker and research into resistance mechanisms.

I’ll be off to San Francisco later this month to attend the AACR meeting on the PI3-kinase/mTor pathway in cancer to see what progress is being made.  Watch this space for updates on what the key opinion leaders think.

References:

ResearchBlogging.orgLovly, C., & Carbone, D. (2011). Lung cancer in 2010: One size does not fit all Nature Reviews Clinical Oncology, 8 (2), 68-70 DOI: 10.1038/nrclinonc.2010.224

2 Comments

"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

One of the interesting things about scientific conferences such as AACR and ASCO is that everyone looks at the same data differently as if it were through a kaleidoscope.

Brand marketers focus on their competition by tumour type or disease, scientists look at specific mechanisms or pathways, investors look at particular companies and so on. 

Someone asked me the other day how I analyse the data.  I hadn't really thought about it much until then, but on reflection what I'm interested in is trends and how research evolves from a big picture science view so that means I look at pathways like a true biochemist.  This also teaches us where the gaps are and what opportunities may arise in the future.  It's not exactly rocket science, but it is a useful approach sometimes.

Phosphoinositide 3-kinasesImage via Wikipedia

One of the clear trends emerging at AACR the other week is that dual inhibition of both the PI3K-mTOR and RAS-ERK pathways may be necessary in some cancers such as melanoma to reduce cross-talk, feedback and feedforward loops, drug resistance and loss of PTEN gain of function, just as one might also target IGF-1R and EGFR to reduce cross-talk and add in another inhibitor, eg MEK or AKT.

Given the increasingly critical role of MEK and AKT in various combinations in the future to reduce the potential for drug resistance occurring, this bodes well for a host of companies.  I wasn't, therefore, surprised to see Novartis snap up Array's MEK inhibitor (ARRY-162) given they already have an mTOR on the market (everolimus, Afinitor), two PI3-kinases in development and others including a RAS inhibitor.  Having a MEK inhibitor as well may therefore give them a lot of flexibility with different combinations in multiple cancer types if this approach pans out. 

Merck are also following a similar approach with their mTOR inhibitor, ridaforolimus, which they have finally grabbed commercial control of from their partner, Ariad.  Let's not forget they also have an AKT inhibitor, dalotuzumab and a MEKi through their partnership with AstraZeneca to play with too.

This is all good news for several biotech companies though, if some big Pharma companies start catching onto the trend and realise they need may a PI3K-mTOR inhibitor and a MEK or AKT inhibitor to stock up in their pipeline before the field gets too crowded.

Which companies might have new and interesting data in this area?

Well, Keryx and Aeterna Zentaris, Semafore, Calistoga, Intellikine and a few others all have PI3K inhibitors in development, while Exelixis have a deal in place with sanofi-aventis for XL147 and XL765 and Roche/Genentech have a pan-PI3K inhibitor, GDC-0941.  Novartis have two (BEZ235 and BKM120). Some of these compounds are single PI3K inhibitors and some are dual inhibitors of PI3K-mTOR.

Looking at the ASCO abstract titles, Exelixis appear to have the most abstracts in this area this year, so it will be interesting to see what sort of data they have across a range of different tumour models and early phase I results in solid and hematologic malignancies, with a variety of different combinations. 

One session I'm really looking forward to at this year's ASCO is a Clinical Science Symposium entitled, "Paths for Clinical Development of PI3K Inhibition" with some of the heavyweights in the field such as Neil Rosen (MSKCC), Skip Burris (Sarah Cannon), Jose Baselga (Spain) and Carlos Arteaga (Vanderbilt).  Arteaga is presenting a talk in that session entitled, "Next steps in clinical development of PI3K inhibitors?"

More later on this blog after the posters and the data become available at the meeting.

Reblog this post [with Zemanta]
1 Comment

There are a lot of clinical trials out there right with tyrosine kinase inhibitors; unfortunately many will fail because they were rushed into phase II or III trials without thinking through all the options.  There are, however, some smart companies out there who do think.

What was noticeable at AACR this year, was the surfeit of posters and presentations regarding logical combinations designed to eliminate escape routes and hence resistance.  For example, cross-talk is a common problem between ligands, eg IGF-1R and EGFR, so combining the two may reduce the problem but that isn't the whole story.

Feedback loops also exist, so targeting PI3-kinase alone is less likely to be effective than targeting both PI3-kinase and mTOR.  Neal Rosen from MSKCC showed some interesting data to this effect and argued cogently that oncogenes tend to lead to constitutive negative feedback.  He also noted that the BRAF mutation predicts for sensitivity to MEKi, for example.  Michael Korn also discussed the feedback activation loop between the RAS-ERK and PI3K pathways and how the inhibition of autophagy (where cells self digest themselves) can enhance apoptosis and the anti-tumour effect with smart combinations.

Targeting both MEK and AKT may therefore also have more effect than either alone, as you can see in the chart below: 

Picture 10Source: Array Biopharma

In a recent trial reported at the the ASCO GI meeting in January, Merck described an elegant design where IGF-1R, EGFR and AKT inhibitors were all combined to target advanced pancreatic cancer, with promising early results.  I thought this was a prescient approach at the time, since it clearly sought to eliminate both cross-talk and feedback, so it was interesting to see numerous researchers advocating similar approaches in different tumour types based on the overexpression profiles at AACR last week. The design is based on rational biochemistry, which regular PSB readers will know I'm a big fan of, rather than randomly adding a kinase inhibitor to whatever is the standard chemotherapy of the day in a haphazard blunderbuss approach.

There are a number of MEKi and AKTi inhibitors out there (I counted nearly a dozen last time I checked), as well as a plethora of PI3-kinase and mTOR inhibitors, either alone or in combination.  Merck and AstraZeneca announced an interesting deal earlier this year to jointly pursue research with their AKT (MK-2206) and MEK (AZD6244) inhibitors.  This collaboration makes a lot of sense biochemically.  Novartis (a client) have one of a broadest kinase pipelines in the industry and just added to it prior to AACR in a deal with Array BioPharma to license their MEK inhibitors, of which ARRY-162 is the lead candidate. 

The compounds that ultimately win the race may not necessarily be the ones furthest ahead in clinical trials right now, but the ones with the smartest clinical trial designs to eliminate some of the issues associated with kinase inhibition – cross-talk, feedback, feed-forward loops and additional mutations. 

MEKi and AKTi are two of my favourite kinase approaches right now because they offer the flexibility to add to existing TKI's such as erlotinib, sorafenib or everolimus, for example, potentially improving the outcomes further in a variety of different cancers, never mind the future combination possibilities.  It's going to be a very interesting and hot area to watch in the near future, that's for sure.

If you have any thoughts or questions on this fascinating topic, please do add them in the comments below.

 

4 Comments

As many readers here on PSB know, I've not been a big fan of genome-wide molecular profiling, preferring an oncogene addiction approach to drug development and targeted cancer therapies. However, every once in a while something comes along that stops you in your tracks and makes you think differently.

This morning I was reading the latest copy of the New England Journal of Medicine over coffee and was fascinated by a review article by Drs Lenz and Staudt at the NCI on the molecular genetics of diffuse large-B-cell lymphomas (DLBCL), which account for 30-40% of newly diagnosed lymphomas.

As the review article points out, it is well know that different subsets of diffuse large-b-cell lymphomas are associated with different overall survival rates after initial anthracycline based therapy.  For example, it is more favourable in people with PMBL and the GCB subytype but less favourable in those with the ABC subtype.  R-CHOP therapy has improved survival in people with ABC, but the cure rates are still lower than those with the GCB subtype.  Gene expression signatures can help identify the subtypes and predict survival rates:

image from content.nejm.orgSource: NEJM

Current therapeutic treatment with chemotherapy has made headway in improved survival, but in order to make further headway, new approaches are very much needed.  Targeted therapies have now begun to expand clinical trial options. 

The article talks about numerous pathways, but I particularly liked this one, which details the Nuclear Factor kB (NFkB) signalling pathways in normal and malignant lymphocytes:

image from content.nejm.org
Source: NEJM

Essentially, signalling is initiated when a SRC family kinase ( SFK) phosphorylates tyrosines in the immunoreceptor tyrosine-based activation motifs (ITAMs) on B-cell subunits.  SYK is then recruited to the ITAMs through the SH2 domains and becomes active.  Many of you will remember SYK inhibition from previous posts on Rigel's fostamatinib, a SYK inhibitor.  I think Celgene also mentioned a SYK inhibitor in early development at their recent R&D Day, although the Rigel-AZ is further ahead but more erratic in it's results, at least in immune disorders.  A more recent paper in Blood looked promising in NHL and CLL, though.  Companies are clearly starting to look at specific inhibitors in the downstream pathway for lymphomas and chronic lymphocytic leukemia.

What's interesting about the cartoon above is that you can also see that phosphatidylinositol-3-kinase (PI3-kinase) is activated in parallel, activating the mTOR pathway.  These two targets are getting a lot of attention from Pharma in the clinic, especially in leukemias and lymphomas, and we may well see more of their latest development at AACR next week and ASCO in June.  Exelixis and their partner, sanofi-aventis (a client), for example, have already announced 12 abstracts at ASCO, including 6 on their PI3K and mTOR inhibitors, but they are focusing on lung cancer, a much more difficult carcinoma, rather than NHL, where there is a strong rationale.

It's good see new treatment modalities being tested in leukemias and lymphomas and not just solid tumours, where most companies inevitably focus due to the larger population sizes.  That said, the challenge in lymphoma is going to be identifying rational combinations that kill lymphoma cells synergistically.  As we learn more about the underlying biology of the disease, targets and biomarkers, so more effective and less toxic solutions may evolve.

ResearchBlogging.org
Lenz, G., & Staudt, L. (2010). Aggressive Lymphomas New England Journal of Medicine, 362 (15), 1417-1429 DOI: 10.1056/NEJMra0807082 

Friedberg, J., Sharman, J., Sweetenham, J., Johnston, P., Vose, J., LaCasce, A., Schaefer-Cutillo, J., De Vos, S., Sinha, R., Leonard, J., Cripe, L., Gregory, S., Sterba, M., Lowe, A., Levy, R., & Shipp, M. (2009). Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia Blood, 115 (13), 2578-2585 DOI: 10.1182/blood-2009-08-236471

error: Content is protected !!