Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts from the ‘Pathways’ category

This month has brought a flurry of regulatory activity in the prostate cancer landscape with several companies seeing noticeable action in the new product development area:

For this specific post, I want to concentrate on the new trends in advanced prostate cancer, as last weekend I attended the 2012 American Urological Association (AUA) annual meeting, focusing on the basic research sessions hosted by the Society of Basic Urological Research (SBUR) and Society of Urologic Oncology (SUO).  It’s hard to ignore the clinical data though, with so much activity in going on in advanced prostate cancer!

While at the oral and poster sessions, I enjoyed informal chats with urologists and researchers. Many of the urologists I spoke to at AUA were very excited about enzalutamide, certainly much more enthusiastic than in the last two years when we saw the arrival of sipuleucel-T (Provenge) and abiraterone (Zytiga) in the pre- and post- chemo settings respectively. The reasons for this were varied, depending on the respondents, ranging from understanding the mechanism of action (MOA) clearly to ease of logistics to lack of concomitant steroid therapy. The fact that the initial overall survival of 4.8 months for enzalutamide in the post-chemo setting, which hints at a more potent and effective therapy, probably helped as well.

With that in mind, I spoke with Dr Neal Shore, one of the AFFIRM trialists about his perspective of the impact of enzalutamide on his patients.  Firstly, we discussed the expectations for patients who have received chemotherapy:

“Patients who typically have completed a course of docetaxel have historically a 6-12 month life expectancy depending upon… on how symptomatic or asymptomatic they are.”

Secondly, what was the impact of treatment with enzalutamide?

“In the treatment arm, those who received enzalatumide, they lived at the median 4.8 months longer than patients who received the control placebo.

So in fact their survival extended beyond 18 months. I told you that normally this patient population has a 6-12 month survival expectancy range, that to me is dramatic.

It’s a dramatic life prolongation effect.”

Emphasis mine.

One of the things that makes enzalutamide exciting for me is the ability to target splice variants. This may explain why the agent has slightly better efficacy in the post chemo setting than abiraterone, which demonstrated a 3.9 month overall survival advantage over placebo when it was initially approved. This figure has since improved to 4.7 months with more mature data, suggesting that the initial 4.8 months improvement we see with enzalutamide may also potentially improve further with time.

Previously, I discussed the splice variants with Dr David Hung, the Medivation CEO:

Dr David Hung on Splice Variants:

Another area where we may see changes in mCRPC is sequencing, combinations and different trial designs. While it seems logical that good old fashioned sequencing, as with ADTs in earlier disease, will prolong time to disease progression by managing PSA levels, it should be noted that I don’t think any of those agents have actually shown a significant improvement in overall survival in patients. Unlike oncology, urology tends to have looser endpoints and this is something that we may well see changes in going forward as more rigour is applied in the clinical trial setting.

Combination therapy is something that I think we will also see more of in the future if we are to see real shifts in meaningful outcomes. As Charles Sawyers noted in a previous interview, shutting down the AR pathway more comprehensively with dual inhibition and ‘big guns’ makes solid scientific sense. This doesn’t just mean the obvious though, such as determining whether dual upstream and downstream AR inhibition with enzalutamide plus abiraterone versus either alone would work, but also targeting cross-talk and adaptive resistance pathways such as AR inhibition plus a PI3K inhibitor. Some of these trials are already underway, at least in phase II to see what the safety and efficacy signals looks like.

The arrival of multiple new therapies that change the standard of care also means that once the current crop of new drugs have been approved (abiraterone, enzalutamide and alpharadin) it becomes more difficult to conduct placebo controlled trials with OS any more, as Dr Shore astutely observed:

Dr Shore’s point about surrogate markers of survival in advanced cancer is a highly relevant one given the increasing level of competition in this tumour type.

In the pre-chemotherapy setting, both abiraterone (302 trial) and enzalatumide (PREVAIL) appear to have progression-free survival (PFS) and OS as co-primary endpoints. It will be interesting to see how the FDA react if only PFS is significant while OS is not, as many commentators suspect from the rather vague and woolly press release that J&J put out for abiraterone.  I don’t know what was agreed beforehand with the FDA regarding the study design, but I can see an interesting and highly charged ODAC ahead here as well as much speculation prior to the ASCO presentation of the data in the prostate cancer oral session next month.

OS is a clear, but challenging measure because ultimately, as Dr Shore noted, the endpoint is death.  Remember, recent breast cancer trials discussed here on PSB achieved a significant OS of 6 months or more, precisely because crossover wasn’t allowed. These trials had an active arm as a comparator, not placebo, though. We don’t yet know if patients in the placebo arm who relapsed were allowed to crossover to abiraterone early (ie before the IDMC recommendation). If they did, then OS was doomed from the start and the trial design itself was flawed, but we will have to wait for the presentation to see before jumping to conclusions.

I really hope that’s not the case here, because once crossover does occur, it is very hard statistically, to sort out a true survival signal. If the placebo patients didn’t crossover before the IDMC recommended study stoppage, then it’s hard to see why OS wasn’t met (if truly the case) unless the patients had long term compliance and adherence problems as a result of the concomitant steroid therapy. I do find it hard to believe, however, that an IDMC would stop a trial early if a primary registration endpoint was not met, for that would be akin to regulatory hari-kiri!

Overall survival has long been the standard of care in advanced prostate cancer and several drugs have either received approval or will receive approval as a result of meeting that high hurdle. Future entrants will find it very hard to ethically justify using placebo in the comparator arm and will likely need to be compared to sipuleucel-T, docetaxel, cabazitaxel or abiraterone as the currently approved new standards of care and potentially other options will be considered that are seeking approval such alpharadin and enzalutamide in the near future.

This improvement in care raises the bar for companies considering advanced prostate cancer in several ways:

  • Increased costs (active comparator arm, prolongation of treatment)
  • Increased time to market (OS takes longer than PFS)

In the end, though, for men with prostate cancer it’s largely all good news as new therapies that clearly prolong life become available and make a difference to their lives, not just in terms of more time with their families, but also in terms of improved quality of life and symptom management.

7 Comments

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

It’s that time of the year again where we cogitate and contemplate on what might be hot at the annual meeting of the American Society of Clinical Oncology (ASCO) before the abstracts are available (they’re released online tomorrow at 6pm ET).

This year, while interesting early data from up and coming small biotechs is likely to be eagerly presented in poster sessions, the focus is more likely going to be on big Pharma with various phase III and also late phase II trials that are due to report data.  Unfortunately, not all of these will produce overwhelmingly positive results though!

What I’m most interested is things that shift the needle meaningfully  in terms of survival by six months or more, as we saw from the recent BOLERO2 and CLEOPATRA trials in ER+ and HER2+ breast cancer.  There are plenty of agents that offer minor or incremental improvements (colon cancer has long suffered from that syndrome, sadly), but let’s be honest – most of us get excited by the possibility of major shifts in survival.

Please note that I’ve mostly selected some promising agents in development that might achieve that effect, explained why they are different and focused on new data/drugs rather than rehash what I call the ‘middlings’ i.e. minor upgrades to the standard of care.

Without much further ado, here are my ASCO preview highlights for 2012:

Please do check back during the convention both here on PSB, and also on Biotech Strategy, for reports and analysis as the interesting data emerges at ASCO.

If you have any comments or thoughts, please do share them below…

4 Comments

At the recent American Association for Cancer Research (AACR) meeting, I had the pleasure of meeting several interesting young scientists and physicians either in the poster halls or in various scientific sessions.  It seemed a great idea to encourage some of them to contribute some guess blog posts here on PSB.

Laura Strong, Quintessence Biosciences

Dr Laura Strong, Photo courtesy of Pieter Droppert, Biotech Strategy Blog

Amongst the people I met was Dr Laura Strong, President and COO of Quintessence Biosciences.

One of the joys of social media is that sometimes you can get to know people a little from online interactions before you actually meet them in real life, making it much easier to walk up and introduce yourself as a ‘warm’ rather than ‘cold’ contact from a conversational standpoint.  I’ve been following Laura (@scientre) for a while on Twitter and was keen to learn more about what her early stage biotech company does.

Quintessence Biosciences is, according to it’s website, “a biopharmaceutical company focused on development of novel protein-based therapeutics as anti-cancer agents. The Company’s products are based on the EVade™ Ribonuclease Technology which allows for the engineering of human proteins (ribonucleases) for the treatment of human diseases.

Essentially, in plain English, this means that “The EVade™ Ribonucleases degrade ribonucleic acids (RNA), resulting in inhibition of protein synthesis and cell death.”

Source: Quintessence Biosciences

Laura was presenting a very interesting poster at the meeting, so I asked her if she was interested in writing a guest post about their work on RNases. She has most kindly agreed, so today and tomorrow we’re running a two-part mini series from Laura on RNases based on Quintessence’s work. For those interested in background research, you can check out more about the company here and also Laura’s blog, The Next Element.


RNases: From Concept to Clinic

At this year’s AACR Annual Meeting, I presented results from in vitro screening of combinations of our clinical stage ribonuclease (RNase). The theme of the meeting, Accelerating Science: Concept to Clinic, captures the serendipitous discovery that started on this course and subsequent development of this innovative and differentiated class of drugs.

Is RNA a good therapeutic target?

RNA has been a validated drug target for decades – from the discovery that various classes of antibiotics target ribosomal RNA to the more recent approaches using modified oligonucleotides to target specific RNA sequences.  Vitravene is an oligonucleotide designed to binds a critical cytomegalovirus (CMV) messenger RNA that was approved by the FDA to treat CMV retinitis in immunocompromised patients.  Recently a New Drug Application (NDA) was recently filed for another oligonucleotide drug, Kynamro (mipomersen sodium) that targets apolipoprotein-B to treat severe forms of familial hypercholesterolemia. These drugs have another feature in common: they do not target cancer.

In cancer drug development, the development of receptor tyrosine kinase inhibitors (TKIs) provides a potential roadmap for successful development of RNA-based therapies. While the early approved drugs, such as imatinib (targets bcr-abl to treat Philadelphia positive Chronic Myeloid Leukemia (CML)), provided significant benefit to patients, resistance via mutation in the ATP-binding pocket of the kinase domain has become a persistent problem in TKI therapy. This situation has prompted the development of second generation drugs (e.g. dasatinib and nilotinib for CML).

Another important lesson from TKI drug development is the clinical impact of targeting multiple and complementary aberrant signaling pathways. Even if the activity of one component of a pathway is blocked, there are often others that can compensate for the loss. In practice, this has led to development of pan-kinase inhibitors and to combining drugs in clinical trials based on the overlap of pathways. These results suggest that a single target approach may not have enough impact in targeting the RNA in cancer cells.

How do you go after multiple RNA targets?

One approach to target multiple RNA sequences inside a cell is to deliver multiple RNA drugs, such as modified oligonucleotides. Alnylam has taken this approach with their early clinical drug ALN-VSP. The drug uses small interfering RNA (siRNA), which are relatively short (1–22 base pair) RNA duplexes that inhibit messenger RNA once inside cells. In the case of ALN-VSP, two types of siRNA are included in a lipid nanoparticle. ALN-VSP targets two genes involved in liver cancer: kinesin spindle protein and vascular endothelial growth factor. The drug has completed a Phase I dose escalation study.

An alternative approach to attack multiple pieces of RNA in cancer cells is to use a human protein whose function is to degrade RNA, a ribonuclease (RNase). While this alternative may not be immediately obvious, serendipity played a role in bringing this concept to the clinic.  In the late 1980s, frog egg extracts were screened for in vitro anti-cancer activity with positive results. The active component turned out to be a frog RNase that is part of the RNase A family.

Professor Ronald Raines at the University of Wisconsin made the connection that bovine RNase A, the prototypical family member, was not toxic to cancer cells and identified a major difference between the frog and bovine RNases. The bovine RNase A binds very tightly to the ribonuclease inhibitor protein found in the cytosol of human cells while the frog RNase does not. Using this information, a variety of bovine RNase A variants were produced with diminished binding to the inhibitor and these RNases were cytotoxic to cancer cells in vitro.

Using the closest human homolog, human RNase I, we first tested the concept of whether certain RNase variants may have anti-tumor activity in preclinical cancer models in mice. Forty human RNase I variants were produced based on data from a crystal structure data of the bound RNase and inhibitor and then tested in xenograft models. The RNases showed a range of activity, highlighting that the activity of the RNase is based on evasion of ribonuclease inhibitor but there are other factors, such as internalization and pharmacokinetics that also contribute to efficacy.

QBI–139

We selected QBI–139 as our lead candidate because the drug had the greatest activity across the most tumor types, including breast, colorectal, non-small cell lung, ovarian, prostate and pancreatic cancers. QBI–139 maintains 95% sequence identity to the human RNase I. The efficacy of QBI–139 was similar to chemotherapies and targeted agents when tested in preclinical models. We also did not see the common toxicities associated with chemotherapy (e.g. myelosuppression, alopecia, etc.) in the efficacy models.

The example shown is a xenograft model of prostate cancer (DU145) comparing QBI–139 to the standard of care agent docetaxel as well as the frog RNase. On a once weekly schedule, QBI–139 provides equivalent efficacy as the other two agents with less toxicity. At this dose, QBI–139 did not cause death (as in the docetaxel arm) or weight loss (as in the frog RNase treatment arm).


Do check back PSB tomorrow for the second part of Laura’s synopsis on RNases, which discusses the clinical aspects and where Quintessence are going with this interesting and novel concept.

The 2012 American Association for Cancer Research (AACR) meeting in Chicago was interesting for several reasons.  While there were no truly ground breaking data such as in previous years as with, for example:

  • vemurafenib in BRAFV600E melanoma
  • vismodegib in basal cell carcinoma (BCC)
  • crizotinib in ALK+ lung cancer

there were a lot of encouraging signs for the future.

What made the meeting exciting for me was the sheer number of new compounds emerging from late preclinical to early phase I – clearly companies are looking to restock their pipelines with the threat of major patent cliffs imminent.  Not everyone is chasing new compounds to license in!  The sheer breadth and depth of the pathways targeted by the new compounds took me a little by surprise.

It was clear from discussions with numerous new product people that while pipeline boards are starting to look a little healthier than of late, many of the new compounds are not yet public, but effort are being made to expedite these into the clinic too. Roche and Novartis have dominated the oncology landscape of the last few years but there were signs of resurgence from old stalwarts such as GSK, Lilly, Pfizer, Amgen and Merck. Such fierce competition is good to see.

Posters from Day 2 at AACR...

The small biotechs were also showcasing some interesting data and over the rest of this week, I’ll be highlighting some of the ones I liked and also explaining some of the new targets and biomarkers that caught my eye.

There was, however, a noticeable dearth of handouts and QR codes (for the PDFs) in the poster sessions this year, necessitating more requests for copies by email, hence the delay in covering the highlights post conference.

I’m pleased to say, however, that many presenters generously shared their poster offline and some of these will be discussed later this week.

Here are some of the topics I’m going to be covering in more detail:

  • Update on the PI3K-AKT-mTOR and RAS-RAF-MEK pathways
  • New targeted agents in late preclinical and phase I development
  • Review of the Science Policy session on regulatory and clinical challenges in new drug approvals
  • Update on new approaches in colorectal, prostate and lung cancers

Check back PSB daily this week for more detailed analyses and insights…

18 Comments

Today marks the kick off for one of my favourite conferences on the oncology-hematology calendar, with the annual meeting of the American Association for Cancer Research (AACR) being held in Chicago.  It’s all about the science and basic research here, although there are clinical sessions, usually on strategy and early emerging phase I/II data.

Wifi is usually pretty good at the AACR annual meeting, although it can be more variable at the smaller meetings.  Like many attendees, wifi permitting, I’ll be tweeting from the conference and blogging some of the interesting highlights over the next few days.

For those interested, you can follow the Twitter chatter using the conference hashtag #aacr from attendees and non-attendees alike, by clicking on the widget below:

AACR have a strong web and social media presence as well as webcasts of sessions (some free, some paid), thanks to the sterling efforts of Ron Vitale and his web team, who do a fantastic job.

They also have iPad/iPhone or Android apps for those interested in looking at the program on the go and an abstract app as well.  After all, what use is a CD-rom of the abstracts if you’re running around McCormick place with a mobile device?  These days, most conferences, I don’t even take a laptop anymore, so a CD is pretty useless with an iPad or tablet!

One of my favourite tools last year was the video app, which offers selections of short interview of some of the presenters giving the highlights of their talks or findings.  This is a great idea for getting key points out on topics of interest.

Huge doorstop abstract/reference books, CD-Roms and flash keys/memory sticks will soon be a thing of the past.  I confess to being a huge big fan of reading the program as a PDF, so that it can be easily read and bookmarked in iBooks, on a plane, bus, or while running round the poster halls.

All of these digital tools are much more practical and user-friendly – tailored towards the typical on-the-go use by attendees and I sincerely wish more conferences would follow AACR’s lead and move over to the web 2.0 world in this fashion.

The conference runs through Wednesday, so do check back daily for the Twitter updates and we’ll be blogging anything interesting either here on PSB (follow @maverickny) or on Biotech Strategy Blog (follow Pieter Droppert, @3NT).

 

1 Comment

Cancer metabolism is fast becoming an area to watch out for in R&D.  Last month I tweeted that I was attending a one day meeting at NY Academy of Sciences on Cancer Metabolism with keynote speakers Drs Lou Cantley and Craig Thompson. Jonathan Mandelbaum (@biotechbaumer) responded saying it looked like a dress rehearsal of another related meeting he was attending the following week. That was too good an opportunity to miss, so I invited Jonathan to consider guest posting a summary of the Keystone event he attended here on Pharma Strategy. I’m delighted to say he kindly took me up on the offer and what follows is Jonathan’s synopsis, including some references he chose to illustrate the key points, most of which are open access.

Jonathan has the honour of being the first formal guest post from an industry scientist, although the first informal one went to Al Lalani, Ph.D of Regeneron, who sent in an excellent and very amusing summary of last year’s ASCO abstracts.  The original guest post was from an industry analyst, Adam Bristol, Ph.D.

I hope to post more guest blogs here from scientists going forward, as they add variety and different perspectives on the evolution of the cancer R&D process.

Jonathan Mandelbaum

To give you all some brief background, Jonathan is currently a postdoctoral fellow at Millennium Pharmaceuticals, Inc. based in Cambridge, MA. He received his Ph.D. in Cellular, Molecular and Biophysical Studies from Columbia University. His thesis focused on understanding the functional consequences of recurrent genetic lesions in human diffuse large B-cell lymphoma. Prior to Columbia University, he obtained his Bachelor of Science from McGill University in Montreal, Canada and spent his summers as a research student at the Campbell Family Institute for Breast Cancer Research within the Ontario Cancer Institute.


The 2012 Keystone Symposia on Cancer and Metabolism was a hot meeting, albeit in the cold mountains of Banff, Canada. Research into how cancer cells rewire their metabolism to cope with increased energetic and biosynthetic demands has been reinvigorated in recent years, in large part due to several prominent researchers that were featured at the meeting, such as Craig Thompson, Lou Cantley, Reuben Shaw and David Sabatini.

Talks and posters from Agios Pharmaceuticals, Inc., Novartis, Pfizer and Millennium Pharmaceuticals, Inc., provided additional insight into how industry is thinking of translating these latest findings into novel therapeutics. Without getting into much detail (since much of the data remains unpublished), I will summarize some main themes of the conference and takeaway thoughts on future directions for drug discovery.

Central to the conference is the observation known as the Warburg effect, identified by Otto Warbug in 1924. Essentially, Warburg saw that cancer cells, despite being flush with oxygen, rely on aerobic glycolysis for their energy needs, resulting in an excess production of lactate. This phenomenon is common to normal proliferating cells, however, cancer cells have deregulated this process in part through activation of various oncogenic pathways (eg. PI3K, RAS-MAPK) and reliance on the M2 isoform of Pyruvate Kinase (PKM2) (see review by Vander Heiden et al., 2009). Many talks focused on how oncogenic activation of these pathways regulates the activity of different metabolic pathways (eg. glycolysis, the pentose phosphate pathway shunt, amino acid metabolism). For example, it’s known that cells transformed with MYC are dependent on glutamine metabolism for their survival (see Wise et al., 2008).

Is this metabolic rewiring simply an epiphenomenon of oncogene activation or is it truly important for tumorigenesis? Interestingly, mutations in several different metabolic enzymes have been found in certain cancers.  Fumarate Hydratase (FH) and Succinate Dehydrogenase (SDH), enzymes important for the TCA cycle, are inactivated by mutations in rare forms of cancer, implicating mitochondrial dysfunction in these tumors.  Additionally, recurrent gain-of-function mutations in isocitrate dehydrogenase (IDH1/2) have been found in a variety of tumor types, such as glioma and AML.  Most recently, PHGDH, an enzyme important for serine metabolism, was suggested to be the target of a locus recurrently amplified in melanoma (Locasale et al., 2011).

Several talks discussed how these mutations might be important for tumorigenesis. For example, FH-deficient renal tumors have a deregulated anti-oxidant response through activation of the Nrf2 transcription factor (Adam et al., 2011).  The authors suggest that increased fumarate in these cancer cells can promote a post-translational modification of proteins called succination, which can affect protein function.  Gain-of-function mutations in IDH1/2 result in excessive production of the metabolite 2-HG (Dang et al., 2009), which can inhibit the activity of histone demethylases, thus affecting the epigenetic regulation of gene expression (Figueroa et al., 2010).  At least for FH and IDH driven tumors, the common thread appears to be that metabolites can play critical roles in regulating a multitude of cellular processes outside of metabolism itself.

How might this basic research be translated into novel therapeutics?

Two obvious drug targets I mentioned, IDH1/2 and PKM2, are in fact drug programs being pursued by Agios Pharmaceuticals, Inc.  IDH1/2 presents an intriguing opportunity as the mutations are gain-of-function; drugs that can specifically target the mutant protein, akin to imatinib and vemurafinib, might be effective cancer therapeutics.

Although I did not discuss this topic, the autophagy pathway was another focus for several talks at the meeting. As this pathway is important for cells to cope with metabolic stress, and cancer cells face challenges such as hypoxia and nutrient deprivation, targeting autophagy might be a beneficial therapeutic strategy for cancer treatment.

Ultimately, understanding how different genetic dependencies in cancer reprograms cells to be specifically reliant on certain metabolic pathways, might provide synthetic lethal opportunities for drugs that target metabolic enzymes in those pathways.

From a drug discovery perspective, the conference highlighted for me two critical challenges going forward:

  1. What therapeutic window might exist for drugs targeting metabolic pathways? Jeff Rathmell emphasized an important observation that activated lymphocytes are dependent on glycolysis, similar to cancer cells, for their survival. Thus, agents targeting glycolysis might very well have immunological side effects.
  2. Many talks highlighted the plastic and redundant nature of metabolic pathways. Inhibiting one pathway can lead to adaptive flux through another pathway, or even drive metabolic enzymatic reactions in reverse, in order to compensate for that initial block. Understanding and overcoming these pathway redundancies (somewhat similar to the current state of signal transduction drug discovery) will be a key challenge going forward for cancer metabolism translational research.

Jonathan Mandelbaum is currently employed at Millennium Pharmaceuticals, Inc. The views expressed in this article are his own opinion and are not shared by his employer.

References:

ResearchBlogging.orgVander Heiden MG, Cantley LC, & Thompson CB (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science (New York, N.Y.), 324 (5930), 1029-33 PMID: 19460998

Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, Nissim I, Daikhin E, Yudkoff M, McMahon SB, & Thompson CB (2008). Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proceedings of the National Academy of Sciences of the United States of America, 105 (48), 18782-7 PMID: 19033189

Locasale JW, Grassian AR, Melman T, Lyssiotis CA, Mattaini KR, Bass AJ, Heffron G, Metallo CM, Muranen T, Sharfi H, Sasaki AT, Anastasiou D, Mullarky E, Vokes NI, Sasaki M, Beroukhim R, Stephanopoulos G, Ligon AH, Meyerson M, Richardson AL, Chin L, Wagner G, Asara JM, Brugge JS, Cantley LC, & Vander Heiden MG (2011). Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nature genetics, 43 (9), 869-74 PMID: 21804546

Adam J, Hatipoglu E, O’Flaherty L, Ternette N, Sahgal N, Lockstone H, Baban D, Nye E, Stamp GW, Wolhuter K, Stevens M, Fischer R, Carmeliet P, Maxwell PH, Pugh CW, Frizzell N, Soga T, Kessler BM, El-Bahrawy M, Ratcliffe PJ, & Pollard PJ (2011). Renal cyst formation in Fh1-deficient mice is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2 signaling. Cancer cell, 20 (4), 524-37 PMID: 22014577

Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, Marks KM, Prins RM, Ward PS, Yen KE, Liau LM, Rabinowitz JD, Cantley LC, Thompson CB, Vander Heiden MG, & Su SM (2009). Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature, 462 (7274), 739-44 PMID: 19935646

Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Löwenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, & Melnick A (2010). Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer cell, 18 (6), 553-67 PMID: 21130701

6 Comments
EAU-2012-Congress-Paris-View-of-Eiffel-Tower-By-NIght

Sights of 2012 EAU Congress

Greetings from the European Association of Urology (EAU) congress in Paris. Despite the grey drizzle typical of Europe in winter, this is actually quite an interesting meeting with lots of poster presentations.

One poster that caught my eye yesterday was from Martin Gleave’s group on clusterin knockdown synergising MDV3100 activity. Previously, we discussed on this blog how inhibiting clusterin with custirsen (OGX-011) potentiated docetaxel. At the AUA meeting last year, the issue of whether the same would happen with MDV3100 was suggested, as you can see in the short video blog.

Clusterin is also known as testosterone-repressed prostate message-2 or TRMP-2, and has been shown by Miyake et al., (2000) to be important in advanced prostate cancer. This is because the treatment of choice in hormone-sensitive disease, androgen deprivation therapy (ADT), can lead to clusterin upregulation, thereby causing castrate resistance.

The group’s latest study at EAU looked at whether clusterin knockdown sensitised MDV3100 activity and evaluated potential mechanisms for how this might work.

The results showed that:

  1. Neither MDV3100 or custirsen alone affected AR levels, but in combination, the AR protein levels were reduced.
  2. The combination synergistically suppressed LNCaP (human prostate cancer cell lines) in vitro and in vivo compared to monotherapy with either alone.
  3. Inhibition of the AR has been shown to activate the PI3K-Akt pathway, but the combination prevented this from occurring.
  4. Dual treatment also increased AR instability via decreased levels of the AR chaperone, FKBP52.
  5. AR degradation occurred with combination therapy via the proteasome, leading to synergistic repression of AR transcription.

While these data offer a very nice and logical preclinical rationale for considering a combination of MDV3100 and custirsen to overcome castrate resistance in advanced disease, we also need to see clinical evidence in advanced prostate cancer before getting too excited. I like the idea scientifically but Oncogenex, the manufacturers of custirsen, have not exactly been swift at moving their previous trials along, as Luke Timmerman noted his Xconomy article last year.

Ultimately, the proof is always in the (clinical) pudding.

References:

ResearchBlogging.orgMiyake H, Nelson C, Rennie PS, & Gleave ME (2000). Testosterone-repressed prostate message-2 is an antiapoptotic gene involved in progression to androgen independence in prostate cancer. Cancer research, 60 (1), 170-6 PMID: 10646870

2 Comments

A couple of recent controversies in the field of angiogenesis have fascinated scientists and clinicians alike, namely:

  • Does VEGF inhibition lead to more aggressive tumours?
  • What drives metastases and invasion?
  • What is the role of tumour hypoxia in this process?

Data was originally presented in glioblastoma by Rubenstein et al., (2000), showing that anti-VEGF antibody treatment prolonged survival, but resulted in increased vascularity caused quite a stir.  Several other groups subsequently demonstrated in preclinical models that VEGF signaling shrinks tumours, but also results in increased invasion and metastases (see Casanovas et al., (2005), Ebos et al., (2009), Paez-Ribes et al., (2009), for examples).

The mechanism for this process, however, remained elusive. A number of factors have been thought to be contributing, including:

  • Vessel pruning
  • Hypoxia
  • Increased expression of c-MET and/or HGF

The corollary of course, is that once we better understand the underlying biology, we can devise strategies to test new agents in clinical trials. The end result would hopefully be improved outcomes for patients undergoing cancer therapy.

Sennino et al., (2012) performed an elegant series of experiments that were published today in Cancer Discovery and sought to understand the roles of VEGF and c-MET signalling in invasion and metastases by using a variety of VEGF and MET inhibitors in transgenic mouse models of pancreatic neuroendocrine tumours. The paper makes for very interesting reading, which I highly recommend.

Here are some of the highlights:

  1. Tumours treated with VEGF inhibitors such as an antibody (#AF-493-NA, R&D Systems) or sunitinib tended to shrink, but were more invasive as defined by irregular tumour border and presence of acinar cells.
  2. Post treatment with VEGF inhibitors, proliferating cells were reduced in the tumour centre compared to control but there were more apoptotic cells compared to the control. This is consistent with what we would expect from anti-angiogenic therapy.
  3. Interestingly, when looking at mesenchymal markers (eg Snail1, N-cadherin, vimentin) there were stronger bands in Western blots after VEGF therapy. EMT activity is usually a sign of invasion and early metastases in the microenvironment.
  4. Tumours treated with anti-VEGF agents had fewer blood vessels than control, again consistent with expectations for anti-VEGF therapy. However, the reduced vascularity was also accompanied by more hypoxia and greater levels of HIF-1a.
  5. c-MET staining was greatest in tumour cells, but not tumour vessels, after VEGF therapy compared with the controls. The latter is reduced as vessel pruning takes place.
  6. Inhibition of c-MET with PF-04217903 and either sunitinib or the anti-VEGF antibody led to reduction in invasion and tumours with smoother contours, but not greater vascular pruning.

Other experiments were performed with both PF-04217903 and crizotinib (MET inhibitors), as well as cabozantinib, a dual inhibitor of MET and VEGF. When both targets were inhibited together, using either cabozantinib or PF-04217903 plus sunitinib, there was a consistent reduction in invasion and metastases. This also increased with tumour hypoxia and c-MET expression.

What does this data mean?

This is the first paper I’ve come across that convincingly suggests that targeting both VEGF and c-MET simultaneously reduces not only tumour size, but also invasion and metastases, thereby overcoming one of the limitations of treatment with VEGF inhibitors alone.

The work also advances our understanding of the anti-angiogenesic process which involves:

“A complex mechanism involving vascular pruning, intratumoral hypoxia, HIF-1a accumulation, and activation of c-MET in tumor cells.”

As a result, the data also suggest the value in combining VEGF and MET inhibitors with a therapy such as cabozantinib (XL184:

“Inhibition of both signaling pathways by XL184 also reduced tumor growth, invasion, and metastases, and prolonged survival.”

Overall, this was a very nicely put together piece of research and expands our understanding of angiogenesis. It also offers insight into how we can improve clinical strategies with combined VEGF and MET inhibition, which I think we will see more off rather than targeting either pathway alone.

Some of these agents are already approved (e.g. bevacizumab, sunitinib, crizotinib), while several others (MetMAB, tivantinib and cabozantinib) are in phase III clinical trials for various tumour types.  It will be interesting to see how dual inhibition develops in the clinic and whether the animal studies can be confirmed in humans.  I do hope so.

References:

ResearchBlogging.orgSennino, B., Ishiguro-Oonuma, T., Wei, Y., Naylor, R., Williamson, C., Bhagwandin, V., Tabruyn, S., You, W., Chapman, H., Christensen, J., Aftab, D., & McDonald, D. (2012). Suppression of Tumor Invasion and Metastasis by Concurrent Inhibition of c-Met and VEGF Signaling in Pancreatic Neuroendocrine Tumors Cancer Discovery DOI: 10.1158/2159-8290.CD-11-0240

Rubenstein JL, Kim J, Ozawa T, Zhang M, Westphal M, Deen DF, & Shuman MA (2000). Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia (New York, N.Y.), 2 (4), 306-14 PMID: 11005565

Casanovas O, Hicklin DJ, Bergers G, & Hanahan D (2005). Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer cell, 8 (4), 299-309 PMID: 16226705

Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, & Kerbel RS (2009). Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer cell, 15 (3), 232-9 PMID: 19249681

Pàez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, Inoue M, Bergers G, Hanahan D, & Casanovas O (2009). Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer cell, 15 (3), 220-31 PMID: 19249680

2 Comments

Every now and then my eye is caught by reports of new fusion genes being found in different cancers.  Often these descriptions involve researchers across multiple laboratories due to the rarity of the target.  Following a discussion on Twitter yesterday, a friend sent me the link to this interesting paper published in Science Translational Medicine.  Naturally, one of the first things that came to mind was ‘is the identified target druggable?’

Source: wikipedia micrograph of epithelioid hemangioendothelioma (from the liver)

Tanas et al., (2011) used deep gene sequencing and conventional cytogenetics to identify two genes involved in chromosomal translocation in epithelioid hemangioendothelioma (EHE), a rare vascular sarcoma that arises out of endothelial cells, namely:

  • WWTR1, WW domain-containing transcription regulator 1 (3q25) and
  • CAMTA1, calmodulin-binding transcription activator 1 (1p36).

The researchers noted that:

“CAMTA1 encodes a transcription factor that is found in all multi- cellular organisms tested and is evolutionarily conserved from Arabidopsis to humans.”

What is its function?

“Little is known about the protein’s function in mammalian cells, but in humans, the gene is expressed almost exclusively within the brain and has been implicated in memory because high amounts of CAMTA1 mRNA have been identified in memory-related regions.”

Now, there are a couple of other things to note:

  1. Not much is known about EHE, as the sarcoma was only described recently by Weiss and Goldblum (2008) in Enzinger and Weiss’s Soft Tissue Tumors. They appear to affect both sexes equally and are not age dependent, appearing in soft tissue, bone and visceral organs such as the liver and lungs.
  2. There are no current treatment options for EHE, other than surgical removal.
  3. Diagnosis of EHE is currently challenging and requires careful histological examination.

The paper is well worth reading for those interested in the challenges of fusion gene isolation, but what particularly struck me was the prevalence of the translocation in EHE compared with other sarcomas – it appears to be very distinctly different, since none of the others evaluated (nearly 30 of them, was found to have the translocation).

The presence of the fusion gene in EHE but not the other sarcomas strongly suggests a role in tumorigenesis, i.e. it’s an oncogene rather than a tumor suppressor gene.

“We anticipate that understanding the mechanism of WWTR1/CAMTA1 oncogenesis will be instrumental toward developing targeted therapy for EHE, for which none currently exists.”

There are some positive things that emerge from this research. In some ways, identification of the fusion gene in EHE may well change diagnosis and treatment options in the future for patients with the disease, much in the same way that imatinib helped to redefine the diagnosis and treatment of another rare sarcoma, gastrointestinal stromal tumours (GIST) in 2002.  Hope, as they say, is always just around the corner.

If any scientists or pharma people following this blog have something in their pipeline that may target the WWTR1/CAMTA1 fusion gene, then please let me know – it would be useful if we could crowdsource potential therapies aimed at oncogenes actively involved in tumorigenesis of rare cancers.

{UPDATE:  Bruce Shriver from the Liddy Shriver Sarcoma Initiative, patient support group since shared this link via Twitter on the background to the disease in plain English and further development of the fusion gene research – please check it out! }

References:

ResearchBlogging.orgTanas, M., Sboner, A., Oliveira, A., Erickson-Johnson, M., Hespelt, J., Hanwright, P., Flanagan, J., Luo, Y., Fenwick, K., Natrajan, R., Mitsopoulos, C., Zvelebil, M., Hoch, B., Weiss, S., Debiec-Rychter, M., Sciot, R., West, R., Lazar, A., Ashworth, A., Reis-Filho, J., Lord, C., Gerstein, M., Rubin, M., & Rubin, B. (2011). Identification of a Disease-Defining Gene Fusion in Epithelioid Hemangioendothelioma Science Translational Medicine, 3 (98), 98-98 DOI: 10.1126/scitranslmed.3002409

error: Content is protected !!