Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts from the ‘Conferences’ category

There were a number of interesting posters at the AACR-NCI-EORTC Molecular Targets meeting today. Specifically, two on metastatic pancreatic cancer caught my eye. You can read about the other one on Millennium/Takeda’s ADC MLN0264 here.

This is an area of high unmet medical need with the fourth highest number of cancer deaths in the US and a median survival of 10 months or less. Even with improvements in the standard of care, it still remains a miserable cancer to get.

Many of you will be aware that KRAS is mutated in 90% of pancreatic cancer cases. As Dr Barry Nelkin (Johns Hopkins) noted today,

“The good news is that we know the target, the bad news is that we haven’t been able to hit it… Yet.”

We do know that KRAS activates three major signaling pathways, namely:
1. RAF/MEK/ERK
2. PI3K/AKT/mTOR
3. RAL.

Oddly, we have a plethora of inhibitors for the first two, but not for RAL. Unfortunately, RAL signaling is critically important in pancreatic cancer.

Dr Nelkin observed that it has been shown that inhibition with CDK5 resulted in the loss of RAL activity in pancreatic cancer cells, as well as reduction in their ability to form tumours and metastasize in vivo. Interestingly, addition of PI3K or MEK inhibitors further decreased the cells transformation.

Dinaciclib (Merck), an inhibitor of CDK1,2,5 and 9 was shown to block RAL activity in pancreatic cancer cells as well as inhibiting xenograft growth and metastasis. In other words, RAL is now druggable!

The researchers at Johns Hopkins showed some nice preclinical evidence that tumour size was reduced in pancreatic cell lines when dinaciclib was combined with either an Akt inhibitor (MK-2206) or an ERK inhibitor (SCH772984).

These results therefore provided a solid rationale for combining a CDK5 inhibitor with one from the PI3K/Akt/mTOR or RAF/MEK/ERK pathways in the clinic. A phase I combination trial of dinaciclib plus MK-2206 is currently enrolling at four centres, including three in the US.

Overall:

I think this is a most interesting trial with a solid rationale that is well worth evaluating in advanced pancreatic cancer. The story though, gets a little interesting. I tweeted to Dr Nelkin’s colleague, Dr Anirban Mitra, who is now Professor of pancreatic cancer research at MD Anderson and learned something rather surprising:

Cytocidal for the uninitiated means they are seeing pancreatic cancer cells being killed.  It’s a short word but it means a lot.

At the poster today, Dr Nelkin confirmed that he had heard rumours to this effect, but stated Merck had at least committed to finishing the phase I trial. It’s not immediately obvious why MK-2206 might be discontinued or on the chopping block, but who knows what will happen if the phase I data turns out to be stunning.  I for one, sincerely hope that they are.

Other companies with a CDK5 and a PI3K/AKT/mTOR inhibitor in their pipeline would do well to watch out for the readout of these results – they could be very interesting indeed.

Next I’ll be off to the European Cancer Congress (ECC) in Amsterdam. This meeting alternates each year between ECCO and ESMO hosting the event at a different European city.

The last couple of years have seen some nice data that missed the ASCO deadline, other years can bring an update of the already familiar ASCO data. I suspect that this year will be one of those events, with updated PD-1 and PD-L1 data.

If you missed my colleague Pieter Droppert’s ECCO highlights yesterday, you can catch them here, including details of the iPad app and abstracts.

In addition, there were other abstracts of interest that caught my eye, including some solid late breakers:

1. T-DM1 for HER2-positive metastatic breast cancer (MBC): Primary results from TH3RESA, a phase 3 study of T-DM1 vs treatment of physician’s choice. H. Wildiers (Belgium) et al.

The study looks at advanced disease in patients who had received at least two prior regimens. This analysis looks like an interim one, given the full study timeframe is over 3.5 years. I’m particularly curious what the physician choices were to compete with Kadcyla and what the 1 year survival curves look like. It’s a wee bit early to hope that they might separate already, certainly I hope they do!

2. Evaluation of everolimus (EVE) in HER2+ advanced breast cancer (BC) with activated PI3K/mTOR pathway: Exploratory biomarker observations from the BOLERO-3 trial. G. Jerusalem (Belgium) et al.

Originally, I thought this had been presented at ASCO, but the biomarker abstract I found actually referred to BOLERO-2, where they noted that “efficacy was greater in patients with low PI3K expression”, which is an odd finding. The BOLERO-3 data from ASCO presented the initial phase III data for the combination of trastuzumab, vinorelbine and everolimus vs trastuzumab and vinorelbine alone in trastuzumab resistant HER2+ advanced breast cancer. This should be an interesting presentation worth attending.

3. FLT1 gene variation as a major determinant of recurrence in stage I-III non-small cell lung cancer. F. Innocenti (USA) et al.

Many of us familiar with FLT3 in leukemia, but FLT1 is an interesting concept with very little data (or drugs) out there. I will be curious to see if this is a druggable target and where this approach might lead.

4. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in metastatic or locally advanced, unresectable melanoma. F. Hodi (USA) et al.

If you look at five year survival curves for advanced melanoma in the literature, it’s historically around 20% or so when patients have received IL-2, which is where I would expect ipilimumab to be. Trials with DTIC have shown a much lower rate, at around 8-9%. Not every patient is suitable for IL-2 though, so we may be seeing similar survival rates irrespective of the immunotherapy given, but with very different safety profiles.

One of my favourite cancer pathways is PI3K-AKT-mTOR. It’s dysregulated in some 80% of cancers yet we haven’t really seen a major breakthrough with these agents in solid tumours outside of the stunning Afinitor data in relapsed ER+ metastatic breast cancer from the BOLERO-2 study presented at ECCO in Stockholm two years ago.

There are many different permutations out there from single to dual inhibitors and also specific isoforms of alpha, beta, delta and gamma.

One of the challenges with targeting PI3K is that it activates feedback loops. Thus inhibiting PI3K in advanced prostate cancer activates the androgen receptor, while single agent use in advanced breast cancer can lead to activation of HER3. In addition, there have been mixed results with biomarkers and specific mutations/tumour suppressors to date such as PIK3CA and PTEN. This increases the complexity tremendously and therefore speaks to more careful trial selection based on inclusion criteria and also logical combinations to try and shut down the compensatory pathway.

I was therefore pleased to see a few trials reporting early phase I/II data in this vein:

P017: Evaluation of tolerability and anti-tumor activity of GDC-0032, a PI3K inhibitor with enhanced activity against PIK3CA mutant tumors, administered to patients with advanced solid tumors.

D. Juric, J.R. Infante, I.E. Krop, C. Kurkjian, M.R. Patel, R.A. Graham, T.R. Wilson, J.Y. Hsu, J. Baselga, D.D. Von Hoff

According to the abstract:

“GDC-0032 is an orally bioavailable, potent, and selective inhibitor of Class I PI3K alpha, delta, and gamma isoforms, with 30-fold less inhibition of the PI3K beta isoform relative to the PI3K alpha isoform.”

Several confirmed partial responses have been reported and further trials will continue:

“GDC-0032 is a next-generation PI3K inhibitor with promising anti-tumor activity observed in patients with PIK3CA mutant tumors. GDC-0032 is being investigated in combination with endocrine therapies such as letrozole and fulvestrant for patients with hormone receptor-positive breast cancer.”

P079: Hyperglycemia in patients treated with the pan-PI3K inhibitor buparlisib (BKM120): characterization, management, and assessment for pharmacodynamics

A. Azaro, J. Rodon, J.F. Vansteenkiste, Y. Ando, T. Doi, D. Mills, C. Sarr, E. Di Tomaso, C. Massacesi, R.W. Naumann

Source: Novartis

Source: Novartis

BKM120 is also an oral pan PI3K inhibitor that does not target mTOR. Aside from the activation feedback loop effects mentioned earlier, PI3K plays a key role in glucose homeostasis. A number of earlier trials with different PI3K and mTOR inhibitors have reported hyperglycemia as a class effect although they have varied in the degree to which the event occurred.

This study highlights the importance of a potential pharmacodynamic marker (C-peptide) in assessing the insulin response and I’m looking forward to seeing more detail in the poster.

P061: Factors predisposing to development of hyperglycaemia in phase 1 studies involving PI3K, mTOR, AKT and mTORC1 and mTORC2 inhibitors

M. Wong, K.H. Khan, K. Rihawi, S. Bodla, B. Amin, K. Shah, D. Morganstein, S.B. Kaye, U. Banerji, L.R. Molife

Related to the topic of hyperglycemia, the Royal Marsden mined their database for PI3K-mTOR trials and looked at factors that might influence the presence of the glucose spike in order to essentially try and predict which patients were more at risk and improve management. While most patients did not require intervention, but in those that did, metformin and insulin were usually preferred. Interestingly, the main factor emerging in this retrospective study was a prior history of diabetes, which is not totally unsurprising. It will be useful to see if these results can be validated in prospective future trials.

P227: Anti-tumour efficacy of the PI3K inhibitor GDC0941, the dual PI3K/mTOR inhibitor GDC0980 and the MEK inhibitor GDC0973 as single agents and in combination in endometrial carcinomas

O. Aslan, A.M. Farrelly, B. Stordal, B.T. Hennessy

Much has be written about the potential for a PI3K and MEK combination in different tumour types, but so far they haven’t proven to be the home run many of us hoped for.

This preclinical paper looks at cell lines to explore potential targets and synergies in endometrial cancer (EC). They concluded,

“Our data suggest that the mutational status of PIK3CA, PTEN and KRAS can be used as biomarkers to select patients for PI3K and RAS/RAF-targeted therapies. Further, the combinations of the PI3K inhibitors GDC0941 and GDC0980 with the MEK inhibitor GDC0973 are promising approaches for the treatment of patients with PIK3CA, PTEN and KRAS-mutated EC.”

Translating data from simple cell lines to complex human bodies does not always predict response given the variable responses seen from patients with mutations and tumour suppressors in clinical trials. I think it will take a while to tease out what defines and predicts a response in each tumour type much in the same way we saw different effects in advanced melanoma when targeting BRAF with sorafenib versus BRAF V600E with vemurafenib or dabrafenib. The devil is in the details.

And finally, an oral presentation with a very different focus in the PI3K related field that I’m really looking forward to hearing:

#1859 PI3KCA mutations and correlation with pCR in the NeoALTTO trial (BIG 01-06)

J. Baselga, I. Majewski, P.G. Nuciforo, H. Eidtmann, E. Holmes, C. Sotiriou, D. Fumagalli, M.C. Diaz Delgado, M. Piccart-Gebhart, R. Bernards

The authors evaluated:

“The influence of PI3K pathway mutations (PIK3CA, KRAS, BRAF, AKT1) on sensitivity to trastuzumab (T), lapatinib (L), or both agents (L+T) in combination in early-stage HER2-positive breast cancer patients enrolled the neoALTTO trial.”

The goal here is to see the presence of any of the mutations were more likely to lead to resistance and enable better selection of therapy for patients. I will update on this study after the presentation.

More detailed posts and synopses will continue from the meeting itself on Biotech Strategy Blog, where we’ll be sharing our insights and analysis daily.

It’s that time of the year when the annual meeting of the American Society of Clinical Oncology (ASCO) hurtles around with alarming speed out of nowhere and everyone in Pharmaland goes, “ASCO, what already? Is it really June?!” Suddenly the month becomes the focus for many frantic hives of activity.

Immunotherapy

The last two years have seen some unprecedented changes in new therapies emerging to treat several different tumour types, both liquid and solid.  One of the new trends that has begun to emerge is the new class of immunotherapy agents called checkpoint regulator inhibitors.  These include:

  • CTLA-4 (ipilimumab)
  • PD-1 (nivolumab and lambrolizumab)
  • PD-L1 (RG7446)
  • OXO-40 inhibitors (more about those in another post).

This year at ASCO brings forth a lot of new data from the four compounds mentioned. In the video preview we have also attempted to explain how these antibodies work and why they are an important development beyond melanoma. There are data in several tumour types including melanoma, RCC and head & neck cancer at Chicago. In the recent thought leader interview with Dr Robert Motzer (MSKCC), he mentioned PD-1 as a hot topic to watch out for in renal cancer. However, I’m particularly looking forward to seeing the lung cancer data, which has the potential to be really stunning.

In this year’s ASCO video preview, we have included some graphics and an MOA video explaining how these immunotherapies are thought to work. Check it out below!

CLL

Another area that I’ve been watching for a while is chronic lymphocytic leukemia (CLL), which has languished a little in the shadow of it’s CML cousin. Not for long though!

There are a lot of exciting developments here beyond Pharmacyclics BTK inhibitor, ibrutinib. These include new CD-20 antibodies such as Roche’s GA-101 (obinutuzumab) and SYK inhibitors (whatever happened to fostamatinib, one of the hematology highlights of the 2010 ASCO?) where Gilead are now developing an early compound, potentially for combining with their PI3K-delta inhibitor, CAL-101, now known as idelalisib.

In addition, Infinity also have a PI3K-delta inhibitor, although they are further behind in development. We don’t know yet whether greater in vivo potency will translate to the clinic or whether also targeting gamma will add to the efficacy or introduce off-target kinase toxicities.  Either way, it’s good to see so many targets and exciting new agents being explored for this disease.

Breast and Lung Cancers

On the solid tumour front, I was delighted to see new data in HER2+ breast cancer and ALK+ lung cancer.  Interestingly, in both of these cancers, Pfizer and Novartis in particular are making inroads with a number of compounds including everolimus (Afinitor), palbociclib (PD-0332991) a selective inhibitor of cyclin dependent kinases (CDK) 4 and 6, LDK378 and PF-05280014, a trastuzumab biosimilar.

Pancreatic Cancer

My final topic that has some interesting developments is pancreatic cancer.  Since the phase III Abraxane data from the MPACT study was presented at ASCO GI, Celgene have filed with the FDA and received Priority review, with a PDUFA date of September 21st.  An update is expected at ASCO, along with tumour marker data and prognostic biomarker data.  Threshold are presenting their phase III study design for TH-302 in the Trials in Progress section, but given the standard of care may well have changed by the time the data is mature, this may well be a day late and dollar short.

All in all, a good year can be expected for new data emerging at this year’s ASCO.

You can learn more about these topics, including insights on how PD-1 and PD-L1 immunotherapies work from the video highlights by clicking on the image below:

ASCO 2013 Preview Video

My ASCO preview video was freely available for several months but is now part of Biotech Strategy Blog Premium Content.

 

2 Comments

One of the hot topics at this year’s annual ASCO meeting is clearly going to be PD-1 and PD-L1 immunotherapies, following on from the success of BMS’ PD-1 agent highlighted in my ASCO video last year.  By now, we know that it has a generic name, nivolumab, and is being studied in combination with ipilimumab (Yervoy) in metastatic melanoma. You can find the many nivolumab abstracts here.

Understanding how PD-1 and PD-L1 immunotherapy works.

Source: Roche

Interestingly, BMS also now has competition from other big pharma companies, including Roche’s PD-L1 antibody, MPDL3280A, and Merck’s lambrolizumab.  Roche appear to be developing a companion diagnostic, which could mean the responses are different for PD-L1 positive and negative patients.  At least, that would be an intuitive conclusion and a potentially good way to preselect patients who are most likely to respond to the therapy.

In addition, the FDA have already picked up on this class of agents, conferring the almost ubiquitous Breakthrough Therapy status on some of them already, so solid data in the next year or so could mean a fast track to market strategy is possible in this category.

In the battle of the abstracts and sheer depth of data in the PD-1/PD-L1 segment, you can see that Merck are already the poor cousins to BMS and Roche in execution. Just one?  That’s pitiful!  Never a good sign.

What factors need to be considered in looking at the immunotherapy data?

Last night I was searching the abstracts on my iPhone after the local broadband inconveniently went down and also watched the conversations on Twitter, usually a fun experience fishing for and discussing the diamonds in the rough. A couple of things struck me, however, around the immunotherapy data mining and chatter:

Firstly, there’s way too much focus on ORR (overall response rate) and the minutiae of the differences between the different PD-1/PD-L1 agents. It’s far too early to tell much, as we all know that what matters are the randomized phase III trials, trial design and patient selection (specific, catch-all etc). These can all have a huge impact on the final outcomes in large scale randomised studies.

Secondly, ORR is a measure of disease control – it tells us how much shrinkage there is going on at the time of measurement and is based on RECIST. This is partly a hangover form old chemo days, and partly a lack of available biomarkers of response. Let’s not also forget that immunotherapies usually have a delayed effect and while waterfall plots at six months or so are useful, they don’t tell us what the long term effects will be.  How durable will the responses be beyond 6-8 months?  Is there adaptive resistance developing?  What sort of logical combinations and sequencing options can be considered? So many questions to which we have no answers yet.

Thirdly, be very careful when interpreting the abstract data for ORR – sometimes the data is given for all the patients, irrespective of whether they responded or not, and sometimes it is given as a percentage of the patients who actually had some sort of response. You need to compare apples with apples when looking across studies or the conclusions drawn can end up being a little off.

Fourthly, I don’t think ORR is the ideal endpoint.  So what?  What really matters is how long do patients live, do they feel better (or worse) and will they have a better quality of life as a result of taking the medication?  Other obvious but important questions we need to evaluate going forward include:

  • How much of a prolonged effect with PD-1/PD-L1 immunotherapies have over 5 years?
  • What will be their effect on subsequent therapies?
  • Will they boost or hinder sequencing and in which tumour types?
  • Is there a biomarker of response?
  • Is a diagnostic necessary?

Fifthly, combination studies are nice if they lead to improved outcomes, but at what cost will this be achieved? By this, I mean both in terms of safety (remember ipi and vemurafenib were thought to be a logical combo in melanoma until the unexpected AEs scuppered that concept) and also cumulative cost of treatment. None of the new oncology therapies can or will be considered inexpensive these days, especially when the benefit might be measured in only a few months or less.

Overall:

I’m really looking forward to these presentations on PD-1/PD-L1 and will write about them in more detail at the meeting.  It continues to be an exciting area in oncology, as long as the results live up to the expectations.  It’s still unclear which tumour types will benefit most and what the durability will be.  Right now, I have more questions than answers, but as a concept it’s definitely one well worth watching over the next couple of years.

2 Comments

On the final day of the annual 2013 meeting of the American Association for Cancer Research (AACR) in Washington DC, Jeffrey Engelman (MGH) hosted an excellent plenary session on “Cancer Evolution and Resistance” with a series of superb talks not only from himself, but also Neal Rosen (MSKCC), Todd Golub (Broad Institute) and René Bernards (Netherlands CI).

If this session is included in the webcast, I would highly recommend watching the whole thing several times, as it was one of the meeting highlights for me. Despite being on the very last day, the large hall was pretty packed and well worth waiting for. You can check availability of the AACR 2013 webcast talks here.

I’m going to focus on some of the specifics in NSCLC from Engelman’s talk for this update.

Where are we in the quest to improve outcomes in lung cancer?

Jeff Engelman, courtesy of MGH

Jeff Engelman, courtesy of MGH

Engelman discussed the basics of what we know about adaptive resistance to TKI therapy in solid tumours – most of them (EGFR and ELM4-ALK in lung, BRAF in melanomas, HER2 in breast, and cKIT in GIST) typically being in the range of 8-11 months, with only GIST seeing an impact for nearly 2 years (20 months).

Thus we can see that the resistance develops over time as mutations and amplifications in the tumour evolve in adaptation to the initial efforts to inhibit the target. Indeed, approx. 50% of EGFR lung cancers develop the T790M mutation, while ~33% of resistant ELM4-ALK cancers show new mutations (e.g. L1196M, G1269A and others).

The development of these changes essentially serves as a way to bypass tracks and and continue to allow downstream signalling of PI3K and MEK to occur, thereby driving growth and cell survival. What then happens is a myriad of other pathways become activated to help drive signalling, for example MET, HER2/HER3, IGF1R etc in EGFR driven cancers and EGFR and cKIT amplification in ALK lung cancers.

As an analogy, think of this process like a road traffic system – if the route into New York from New Jersey was cut off at the Holland Tunnel, so traffic would increase to the Lincoln Tunnel or Verrazano Bridge and if those were cut off, traffic would then flow onto the George Washington Bridge, as it adapts and seeks new escape routes from the original destination.

Eventually, the cancer evolves further with defects in growth arrest and apoptosis, as seen with transformation from NSCLC to SCLC in some patients with EGFR cancers, and even changes in the microenvironment through epithelial mesenchymal transition (EMT) and loss of BIM.

The key question is what can we do about overcoming or delaying resistance?

One strategy would be to evaluate more potent inhibitors e.g. LDK378 instead of crizotinib in ELM4-ALK cancers. Another might be to explore logical combinations to address shutting down the bypass tracks. A third might be to add in a new inhibitor to target the specific mutations that evolve e.g. T790M inhibitor in the case of EGFR driven cancers when it appears.

Some of these trials are already underway and we should have more data soon.

Another way, as we saw with the last post on metastatic melanoma, is to identify mechanisms of resistance using laboratory models and lab specimens. This approach can potentially lead to more rational drug development in the clinic. Traditionally, scientists have induced resistance in mice, looked for the mechanisms (a process that can take 1-2 years), validated them in lab samples of patients, and then treated with a new treatment strategy.

This process is obviously time consuming though and not every patient can wait that long for the answer. Engelman then explained how they are looking at ways to streamline the process in Boston. After the mouse resistance experiments are completed, they have added in a drug combination screen to look for logical treatment strategies i.e. what can be added to the original drug to overcome resistance?

A very elegant example was given for EGFR lung cancer where they evaluated 78 test drugs in a screen with and without gefitinib to determine those which led to cell death. Other examples were given for ELM4-ALK cancers.

The screen results suggested that most of the resistant models produced 3-6 hits. These might include adding a MET inhibitor to an EGFR inhibitor in EGFR mutant cancer, an EGFR inhibitor in MET amplified cancers and a SFK inhibitor in the case of ELM4-ALK cancers, for example.

These results are still early, but they do look very promising. Validation studies are still needed, but early studies they performed suggested that the hits are indeed showing efficacy in vivo.  A preclinical example for this concept was shown in vivo by adding ABT-263 (Bcl2 inhibitor) to gefitinib and seeing first a rise in tumour growth with the EGFRi and then a large drop in volume when either ABT-263 or AZD6244 (MEK) was added.

Based on the exciting initial concepts in animals, they are now moving to patient derived models since next generation sequencing (NGS) can help identify the mechanisms of resistance and combined with the drug combination screens, we may see more individual level treatments for patients on a case by case basis.  These might be based on large scale (over 100 cell lines) testing derived from resistant biopsies to identify effective combinations and match them to the relevant biomarkers.  It sounds easy and obvious, but few centres are doing this in practice.

This is true personalised medicine in action.

It is also pretty exciting to me as we know that cancer, even in different patient tumours, is very heterogeneous and requires a more personalised rather than a one size fits all approach. As Engelman observed,

“Heterogeneity of resistant clones within individual patients may pose additional challenges to overcome resistance.”

The second half of his really excellent talk focused on the use of sequential biopsies in patients to explain the heterogeneity and how it can lead to transformation from NSCLC to SCLC and back again in response to treatment with an EGFR inhibitor. That’s an in-depth discussion for another day though, but suffice to say it was a fascinating topic.

And finally…

I can see these novel and applied techniques eventually moving very fast and adopted in top level Academic centres where they have the resources and knowledge to marry basic and translational research with clinical practice in early stage trials, but for many Community or even regional Academic physicians, this will be virtually impossible without referral of patients to clinical trials in the Academic centres, at least for now.

Ultimately, we will see more improvements in treatment for lung cancer when we figure out not only the targets, but also how to overcome adaptive resistance, add logical new combinations, and select future treatment based on biopsies as the tumour evolves its response to each line of therapy. Treatment will essentially need to be chosen on an individual patient basis in the long run by evaluating adaptive resistance to each new combination over time.

The idea that we can use mouse models and drug combination screens with sequential patient biopsies to better understand the adaptive response to therapy over time is not new but few have managed to put processes and strategies in place to make this happen in real time. Patients often can’t wait 2 years or more for a new combination trial to open, but the Boston approach is very promising and I’d like to applaud all those at the Boston group (MGH, Dana-Farber, MIT/Broad etc) for their groundbreaking work in this field. Keep your eyes peeled for more updates in this exciting area of research!

One of the interesting themes for that emerged for me at AACR this year was the amount of effort that is being expended on strategies to overcome drug resistance. This was particularly noticeable in metastatic melanoma and non-small cell lung cancer (NSCLC).  More on lung cancer in another post, as today I want to focus on melanoma.

In the advanced melanoma, vemurafenib is given to patients with the BRAFV600E mutation, which occurs in approximately 50% of patients. This oncogene drives activity of the tumour, but inhibition with vemurafenib (Zelboraf) has shown some remarkable effects, as the stunning before and after photos from Levi Garraway’s group demonstrate.

The challenge, however, is that adaptive or acquired resistance can occur in response to treatment and patients sadly find their melanoma returning after approximately 6-9 months on continuous daily therapy.

At last year’s ASCO, we saw that adding a MEK inhibitor such as trametinib to a BRAF inhibitor such as dabrafenib added around an extra 3 months over single agent BRAF therapy before the resistance set in and the disease returned, sometimes with a deadly vengeance.

I’ve written here on this blog about numerous mechanisms of resistance in advanced melanoma from MEK to COT and others (see related posts below for background reading).

The big question at this AACR was how is the field progressing with new research?

Sometimes, we have to go back to the lab to study animal models of resistance before returning to the clinic with new ideas.

Screen Shot 2013-04-15 at 2.54.50 PMThat’s what a young Novartis Postdoctoral fellow from NIBR, Meghna Das Thakur, did. She asked critical questions and attempted to answer them in a series of elegant experiments with mice as well as retrospectively test the concept in patient data. The cool thing is that while many of the oral sessions were taken up by the Major League researchers is that it’s also nice to see up and coming young scientists present some nicely done research.

What Dr Thakur did was really interesting…

Her hypothesis was simple – that resistant tumours are ‘less fit’ than sensitive cells and have a selective disadvantage over sensitive cells in the absence of drug.

If this were true then we would expect to see dependence on drug for growth of resistant tumours in xenografts, much as we do in humans, with vemurafenib. The data clearly showed that vemurafenib resistant tumours were dependent on drug for growth. They also observed that:

1) p-ERK levels increased following withdrawal of vemurafenib in resistant tumours
2) There was a great deal of heterogeneity in the mechanisms of resistance

The key question then becomes how can we use this information to prevent resistance?

One way to explore this is to look at selective pressure, since vemurafenib is usually given continuously…

  • Treating continuously means that selective pressure enriches resistant cells
  • However, withdrawing the drug means that resistant cells suffer a fitness deficit

Essentially, the researchers noted that, “alternating the selective pressure prevents the emergence of a resistant population.”

In comparing continuous vs intermittent dosing, two things could be concluded from the model:

1) Resistant tumours emerge more rapidly under continuous dosing with vemurafenib
2) Intermittent dosing in multiple tumor xenograft models forestalled resistance

What can we learn from retrospective patient data?

What they did next was to look at computed tomography (CT) scans analysed for evidence of vemurafenib-dependence in patients treated with vemurafenib in the BRIM-3 and vemurafenib safety study, which were identified from the clinical trials database at the Royal Marsden Hospital. In particular, they focused on patients who stopped treatment because of progressive disease (PD).

Of 42 patients identified, 19 of these had CT scans performed after cessation of vemurafenib available for review, while 23 patients did not have a CT available for review and in 16 patients this was because of rapid PD.

For each of those 19 patients with a post-vemurafenib CT, the total tumour volume on three consecutive CT scans was recorded:

  • The CT performed prior to stopping vemurafenib
  • The CT performed when progressive disease was diagnosed and vemurafenib stopped
  • The CT performed after vemurafenib was stopped and prior to the initiation of further therapy

They found that 14 of the 19 patients experienced decreased tumour growth rate following cessation of vemurafenib, supporting the hypothesis that resistant tumours emerge more rapidly under continuous dosing with vemurafenib.

In this model, it is clear that intermittent dosing prolongs survival, but what is the underlying mechanism, and what does this mean for future treatment of patients and also clinical trial designs?

From this initial work it was clear that the MOA is not yet fully understood and further bench work will be needed to elucidate the mechanisms involved.  We don’t yet know, as the researchers point out, whether:

“Does resistance come from a clonal population or is it an adaptation or re-wiring of a selected few cells?”

What was clearer though, is that new clinical research will be needed to evaluate the potential for intermittent vs continuous dosing in patients, particularly in combination:

  • Will there be greater selective pressure from BRAF + MEK inhibitor combinations vs BRAF alone?
  • Identify combination partners that could be used during BRAF inhibitor holiday.

Overall…

I thought this was a very nice piece of well thought out research, perhaps one of my favourites from the AACR meeting this year.  Critically, we saw that the preclinical mouse xenograft model predicted a clinically-relevant resistance mechanism. Removal of drug from resistant cells leads to MAPK pathway hyper-activation and resistant cells experience a fitness deficit. Cycling the mice on and off therapy led to better outcomes than if they were dosed continuously, thereby giving us a new strategy to consider in melanoma patients.

The main impact of this preclinical study is that future clinical research in advanced melanoma should investigate the value of intermittent dosing strategies to improve patient outcomes.

The work turns the old concept of continuous dosing 365/24 on its head – suppressing the BRAFV600E oncogene continuously in melanoma may not necessarily be the best strategy in terms of superior outcomes.

This this does NOT mean that ALL tumours will behave in a similar fashion and intermittent dosing should be tested first in clinical trials where there is sufficient scientific evidence to warrant it.  If I were a patient, considering drug holidays without any evidence of effectiveness would NOT be a good idea.

I’m really looking forward to seeing the results of future combination trials with intermittent dosing to see if outcomes are indeed improved beyond would we currently see with continuous dosing either alone or in combination.

 

4 Comments

This year’s American Association for Cancer Research (AACR) annual meeting grew by 8% to approximately 18,000 attendees with 25% from 75 foreign countries, it is truly becoming a more global event for cancer researchers.

Over the next few days I plan to cover some of my highlights (basic, translational and clinical) in depth here on the blog and also with additional notes for email subscribers.  If you haven’t signed up for the PSB email alerts, there’s still time before the AACR notes go out.

Cherry Blossom in Washington DC by the Monument during AACR 2013With around 6,000 posters and many oral presentations from leading researchers, there is usually some interesting early data coming out from AACR.  This year was no exception.  My pile of poster handouts is over 6” thick with more already coming in my email!  My fervent wish for next year is that more scientists take to the QR code method of sharing their posters – aside from being green and saving trees, it’s also considerably easier on the back!  Another welcome development would be putting the posters online for later download as many of the European meetings already do.

I’m a little tired today as the event only just finished yesterday with a very good plenary session involving Jeff Engelman (MGH), Neal Rosen (MSKCC), Todd Golub (Broad Institute) and René Bernards (Netherlands Cancer Institute).  More on this later, but what a way to end the meeting with a fairly packed hall despite it being the last day.

One of my favourite activities at AACR is talking with young researchers in the poster hall, and a few of these will be highlighted in separate posts.  Many took the time to explain some complex biology and answer my many questions on a variety of topics.  Some of this information was really helpful in improving my own understanding of why I don’t like some therapeutic approaches (e.g. targeting hypoxia) others reinforced my enthusiasm for some immunotherapies such as PD-1 and PD-L1 inhibition.

What about the emergent themes from this year’s AACR meeting?

Every year brings new developments in some shape or form, but here are some of the trends I observed based on the posters and oral sessions I attended:

  • Identifying and developing strategies for overcoming resistance was MUCH more noticeable this year
  • New combination strategies (including more novel-novel approaches) was also very much to the fore
  • Increased pace of research into biomarker identification for clinical trial design
  • Continuing rise of epigenetics as a viable approach for cancer therapeutics
  • New targets emerging (more about these later)
  • Second generation agents to CDK 4/6 and 7, chimeric antigen receptor technology (CART), Polo-like Kinase (PLK1) and many others.

Over the next few days I’ll be writing more about these topics after wading through my many pages of chicken scratch notes from the oral sessions (largely driven by ones I know likely won’t be on the webcast, which goes live for the majority of sessions on May 1st) and that huge poster pile – watch this space!

 

 

After highlighting the interesting biomarker program associated with AVEO’s tivozanib in renal cell and triple negative breast cancers in the last post, several people wrote in asking about other biomarker programs that have piqued my interest. Regular PSB readers will know that I’m not a fan of catch-all trials at all because the population being studied is too heterogeneous – use of biomarkers can help select which patients are more likely to respond to a particular drug and thus produce greater efficacy.

Another small biotech doing some interesting and compelling biomarker work is Array BioPharma, based in Boulder, Colorado.

What’s exciting about the ARRAY pipeline?

They have a nice pipeline of interesting targeted agents that are being developed with several big Pharma partners including AstraZeneca (selumetinib) and Novartis MEK162), both MEK inhibitors. Dennis Slamon’s lab has published some initial data on their work on predictive biomarkers with selumetinib for those interested (see References below).

MEK inhibitors are thought to target KRAS, which causes resistance in lung cancer and melanoma.  Combining MEK with a PI3K inhibitor may reduce the adaptive resistance and prolong survival. I’m expecting to see more on these at the forthcoming AACR and ASCO meetings in April and May. The approach is most likely to be incremental rather than a home run, though.

The ARRAY biomarker program that has intrigued me was presented in multiple myeloma (MM) at ASH in December. They have a novel kinase that targets Kinesin Spindle Protein (KSP) codenamed ARRY-520, which has a novel mechanism of action. For those of you interested in learning more about the basic biology of the KSP concept, I highly recommend checking out the papers in the Reference section below as they explain the targeting of the microtubilin concept well and how KSP inhibition differs from what we know of the taxanes as well as some useful background on the compound itself.

Some of you will remember previous KSPs that failed in the clinic such as ispinesib, for example, but not all compounds are equal or destined to fail. Much of cancer research is iterative – tweaking molecules to improve the conformation (shape), reducing side effects or improving potency. Sometimes a whole program can be canned because the company selected the wrong tumour type or trial design – you don’t always get multiple shots on goal in small biotechs!

What’s interesting to me is the focus of Array in multiple myeloma, a particularly difficult and foxy disease to impact clinically. The basic rationale behind this approach is that KSP is a microtubule protein required for mitosis (cell division); inhibition leads to cell death (apoptosis).

Now, remember the bedrock of therapy in MM has been proteasome inhibitors (bortezomib and now carfilzomib have been approved) and Immune Mediated inflammatory Disease agents or IMiDs (thalidomide, lenalidomide and now pomalidomide are all approved). This means that new agents with a different mechanism of action attract a lot of attention, especially if they can potentially be combined with existing drug classes to prolong survival and push patients into minimal residual disease (MRD) where the myeloma clone is drastically suppressed, much as we see in CML with the BCR-ABL inhibitors.

Inevitably, new drugs in MM are tested in a highly refractive population, either as a single agent or in combination with dexamethasone (dex) to determine if they have any efficacy.

Recall that two new agents approved by the FDA in relapsed/refractory MM had relative low single digit response rates (RR) as single agents i.e. carfilzomib (22.9%) and pomalidomide (7.4%) with improved RR in combination with low dose dex i.e. carfilzomib (34%) and pomalidomide (29.2%)

The two ARRY-520 analyses at ASH looked promising

Dr Satin Shah, ASH 2012

Dr Satin Shah, ASH 2012

The first part of the phase II study from Shah et al., (Abstract #449) looked at patients who were highly refractive to bortezomib and/or lenalidomide.

They observed that ARRY-520 had encouraging efficacy with and without dex. The first cohort (n=32) evaluated single agent therapy with ARRY-520 and saw 19% confirmed responses (CR) and 15% partial responses (PR); the subset who were both bortezomib and lenalidomide refractory had an ORR of 15%.

Meanwhile, the second cohort (n=18) looked at the combination with low-dose dex and demonstrated a PR or better of 22% in a heavily pre-treated population (more than 10 prior regimens). Adverse events included the usual myelosuppression (neutropenia, thrombocytopenia, anemia) seen in myeloma, but importantly, no treatment related neuropathy was observed.

The second element of the analysis looked at the same patients, but described the results from a biomarker, alpha-1-acidic glycoprotein (AAG). AAG is a serum protein that can have elevated levels in MM. As Dr Shah noted, AAG is not known to bind to standard of care agents in MM but is thought to bind to ARRY-520 with a negative impact by reducing the available amount of drug. In other words, they have a sub-therapeutic exposure to ARRY-520.

In the phase II study, they found that the hypothesis was supported: patients with high levels of AAG had poorer responses, while patients without elevated AAG levels had much better responses and the ORR increased to 33%, a dramatic improvement. The pre-dose AAG levels therefore correlated with response.

Obviously, the biomarker will need to be validated in larger, randomized controlled trials, but it would be very useful to be able to select patients upfront who could receive ARRY-520 either in combination with low dose dex or with a proteasome inhibitor or IMiD and see a more pronounced response. An initial trial with carfilzomib suggested an acceptable toxicity profile, while this is an encouraging start, we still need more data on the safety and efficacy of the combinations going forward.

Assuming the ongoing Phase Ib combination trials demonstrate good tolerability and efficacy, ARRY-520 could be potentially be combined with dexamethasone and either carfilzomib or pomalidomide in the relapsed/refractory setting for greater responses than the doublets alone.

Some additional thoughts…

I thought the Array KSP compound looked very encouraging indeed – multiple myeloma is crying out for:

  1. New agents with a different mechanism of action from the existing standards of care that can be combined to give solid results from triple combinations.
  2. More competition in the refractory setting to push out the MOS further and try to achieve minimal residue disease (MRD), which would impact the lives of patients with multiple myeloma significantly.

Beyond Kyprolis and Pomalyst, here are other agents in phase III studies being tested in refractory MM such as the HDACs e.g. panobinostat and vorinostat. The vorinostat data presented at ASH in 2011 was singularly disappointing, but hopefully we will hear about the phase III panobinostat results later this year.

In the meantime, Array have a nice compound in ARRY-520 and a potentially useful biomarker of response to help select patients upfront who are more likely to respond to treatment. As far as I know, they don’t yet have a partner for the program and may well need one for the large phase III trials that will be needed for FDA approval or they may well try to go it alone using the capital raised from the MEK partnerships. MM is certainly a promising avenue worth exploring for ARRY-520 and I look forward to hearing more about its development.

References:

ResearchBlogging.orgGaron, E., Finn, R., Hosmer, W., Dering, J., Ginther, C., Adhami, S., Kamranpour, N., Pitts, S., Desai, A., Elashoff, D., French, T., Smith, P., & Slamon, D. (2010). Identification of Common Predictive Markers of In vitro Response to the Mek Inhibitor Selumetinib (AZD6244; ARRY-142886) in Human Breast Cancer and Non-Small Cell Lung Cancer Cell Lines Molecular Cancer Therapeutics, 9 (7), 1985-1994 DOI: 10.1158/1535-7163.MCT-10-0037

Sarli, V., & Giannis, A. (2008). Targeting the Kinesin Spindle Protein: Basic Principles and Clinical Implications Clinical Cancer Research, 14 (23), 7583-7587 DOI: 10.1158/1078-0432.CCR-08-0120

Tunquist, B., Woessner, R., & Walker, D. (2010). Mcl-1 Stability Determines Mitotic Cell Fate of Human Multiple Myeloma Tumor Cells Treated with the Kinesin Spindle Protein Inhibitor ARRY-520 Molecular Cancer Therapeutics, 9 (7), 2046-2056 DOI: 10.1158/1535-7163.MCT-10-0033

Following last weeks post on the phase III clinical data for tivozanib in advanced renal cell cancer (RCC), I thought it would be useful to provide an update on AVEO’s biomarker program.

I’m very excited about the work they are doing in this area and have been following them keenly since they first presented their initial work on myeloid cells in RCC at the AACR diagnostic conference back in 2010.  Since then, other companies have also published work in this field, including Regeneron, who also noticed the presence of myeloid cells in their work with aflibercept in glioblastoma.

Biomarkers have been very much a bête noire in angiogenesis research – we know that some patients respond well to therapy, but others do poorly and may actually be worse off.  The challenge has been finding a way to link biology with response in order to improve patient selection.  Genentech/Roche have laudably spent millions in their quest for biomarkers with bevacizumab (Avastin), with researchers clearly very frustrated at the confounding data presented at the 2011 ECCO meeting in Stockholm. Clearly, we have a long way to go in this field.

Murray Robinson, AVEO

Murray Robinson, Courtesy of AVEO

Last week I caught up with AVEO’s Murray Robinson (who is the Special Advisor to the CEO), to discuss the progress they are making on the biomarker front.

The recent ASCO GU meeting in Orlando highlighted very interesting data on a new biomarker they are researching, which involves a hypoxia gene signature in RCC (PDF download).

While this data is still preliminary at the moment, it is being tested prospectively in some ongoing trials in both renal and breast cancers.  Readouts are expected by the end of this year and may reveal some interesting findings.

PSB: Does the hypoxia biomarker in your poster at ASCO GU connect with the myeloid biomarker you presented back in Denver at AACR, or is this completely different?

Murray Robinson: This is one is independent of that, completely different, although as you can imagine we are looking at the myeloid biology in this same dataset. To put that to rest, we ran into some technical details, technical challenges on that. Remember when you are doing these trials, you only get the material you get, and you don’t always get the quality, and can’t go back and recut it.

We are really looking forward for the marker in this paper as well as that that myeloid work, to an ongoing study of a 100 patients mostly in the US, RCC – BATON (Biomarker Assessment of Tivozanib ONcology) study and we will get to take another look at this hypoxia biomarker, the myeloid biomarker and a few others that we are looking at. That study will mature soon, we are expecting probably sometime later this year.

PSB: Are these two biomarkers going to be practical in the future for community oncologists to use or is it going to be something that is a research tool do you think?

All of us are now applying high resolution molecular analysis to all of the tumors, all the TCGA work, whole genome transcriptome on things. As the field has done that, we have all been amazed and enlightened by the complexity within what the field used to call a single genotype.

We are all recognizing there are many molecular subsets. This of course was first elucidated 10 years ago in breast cancer. In breast cancer we are very comfortable with these idea, it’s not breast cancer, it’s ER+, or HER2 amplified or triple negative. And of course treatment assignment can be based on that, and we are even further refining those subsets. We haven’t that kind of resolution until recently with many of the other tumor types.

Let’s turn to renal cell carcinoma, (RCC) which is the subject of this work. We in the field have long considered clear cell renal carcinoma, which is 85% of RCC, to be a tumor type. However, as the field has begun to sequence and do profiling on many of these tumors, we have begun to recognize that it is in fact more complex than that.

In our poster, we first cited some great pioneering work by Dr Kim Rathmell down at UNC Chapel Hill where she really was one of the first to take a look at these subtypes of clear cell, and has reported now, and we excerpted one of her figures from a recent paper of hers on our poster. And she sees three major subtypes of clear cell kidney cancer. Two of those subtypes she has talked about before, and we won’t spend much more time talking today. They don’t seem to differentiate for a number of key biological features. A third type she has identified, and we also independently identified, is the subject of this work.

What we, and others, have recognized is that there are new, previously underappreciated, subtypes of clear cell carcinoma.  We focused on this one novel subtype, which Kim called Cluster 3, and we then applied some of our own bioinformatics platform to this, in which we have comprehensively charaterized tumors for key robust signatures.

We applied those key robust signatures that usually represent biology to this third tumor type.  We saw that there were a number of key features in which these tumors varied, this is about 15% of clear cell RCC, so it is a relatively small subset. We noticed in particular that one of the dominant differentiating features of this tumor was the expression of its hypoxia responsive genes.

We recognize there’s a different tumor type here, and that the tumor type is now low for a large signature that we have had for hypoxia, and we of course are thinking translationally, and in terms of molecular diagnostics as well.  We optimized to reduce that hypoxia gene signature to a nine gene signature and we optimized the ability to measure those genes often using single sections from clinical paraffin embedded material.

We are absolutely thinking about this biology and are looking toward to the development of a simple diagnostic test, if that were warranted.

We looked at that nine gene signature, quantified it then looked at its impact on response to tivozanib as well as sorafenib.

PSB: My understanding from looking at your poster in this particular subset with the hypoxic element is that some patients did better in response to tivozanib than others?

Murray Robinson: That is the observation that we made.

Our interest in looking at hypoxia in tivozanib is that hypoxia as we showed in one of our figures, that signature relates to the over-expresssion of key VEGF ligands, VEGFA, PLGF and VEGFC.

One hypothesis that we generated from these observations is that when the VEGF pathway is deregulated or overactive as it is in most of clear cell, then you are more likely to respond to a selective, potent VEGF pathway inhibitor like tivozanib. Conversely, then if you find a subtype that is not, the hypothesis is that it would not respond well.

We tested that in the study, and with the important caveat that we had 69 samples out of the total trial of 517 patients, then we looked at this subtype. Well, we predicted the subtype would represent about 15% of the total, so we went form 69 down to about 30 samples, then 15% of those I think we ended up with 4 of the low subtype in both [arms]. We all recognize those are pretty small numbers. We are really looking forward to the second study I mentioned to you.

PSB: You will still have to validate the results in a larger sample?

Murray Robinson: We think that would be prudent. We think it is difficult to make much out of 4 samples, however that is the amount that would be predicted. It is consistent with all the work that we have done previously. We are also certainly pleased that those 4 patients did not perform well on tivozanib, whereas in the remaining 85% of those patients, those patients did very well.

In fact median PFS of the high patients, representing 85% of the study population, was 18.3 months, which is very respectable progression free survival.

PSB: In other words you have a very good way upfront to potentially predict up front which patients might do well on tivozanib?

Murray Robinson: That was the hypothesis going into this, this was a prospectively designed hypothesis that we wrote up. It was not exploratory.

There is another point here that we’re pretty excited about. One thing we know about the field, that we didn’t 10 years ago, is that despite the original enthusiasm around the VEGF pathway as being a key angiogenesis inhibitor pathway, we all know the large amount of clinical data over the last 10 years with different VEGF pathway inhibitors has been disappointing.

I think there has been a lot of disappointment about the expectations about the VEGF inhibitors versus their performance. The VEGF pathway inhibitors are working, there are a number of indications there, but it certainly hasn’t been that universal broad-acting, cytotoxic agent that we all thought it was, would have been back in 1999.

The bigger question is can we use an understanding of the biology, an understanding of gene signatures, to better select patients in other indications? What we showed in this paper is that 85% of these patients have this VEGF deregulation signature and that is consistent with the fact that clear cell carcinoma actually exhibits robust single agent response to tivozanib and activity with other VEGF type agents.

We really are interested with the signature in expanding and using this as a biomarker outside of clear cell carcinoma, and we have now have done that. Those studies are in progress.

We did a survey of this hypoxia signature across multiple tumor types and made an observation that we are pretty excited about. That is, we found that the hypoxic signature is not as prevalent in those tumor types as it is in clear cell.

We did find that it was present in subsets of tumor types, and we particularly noted that in triple negative breast cancer (TNBC), the hypoxia gene signature is high in a high proportion of triple negative breast cancer.

That is interesting because there is some anecdotal or retrospective clinical data that suggests that VEGF pathway inhibitor agents may work a little bit better in triple negative breast cancer, a challenging tumor type with high unmet medical need.

That observation led to us starting a new phase 2 trial in breast cancer, the BATON-breast study that we started a few months ago. In that study, we will be looking at triple negative breast cancer and we are looking at a randomized phase 2, and we are looking at paclitaxel versus paclitaxel plus tivozanib. In that study we will be measuring the hypoxia biomarker signature.

We have started that and are planning on using a specific assay to look at those patients.

PSB: And finally, for the prospective renal trial with biomarkers when do you expect that to read out, is it anytime soon?

Murray Robinson: I would expect to look for that around the end of the year, recognizing that we don’t know when that’s going to mature, but second half of the year that is when we are expecting. We are looking forward to those results.

We think that there is a strong hypothesis for this and we are pleased in the TIV0-1 study that we saw data consistent with the hypothesis.

Some personal thoughts and additional insights… 

Before anyone gets in too much of a tizzy, note that the data reported at ASCO GU are preliminary findings in a small sample size with only a few (n=4) hypoxia low patients, as Dr Robinson fairly pointed out.  They need to be validated prospectively in a randomised controlled study to ensure that the initial findings are real and not a fluke. The good news, however, is that the consistent lack of response in these patients enabled the hypothesis to be tested further in new trials. It’s certainly a concept that worth testing.

We have known, or suspect, that VEGF inhibitors work in conditions of tumour hypoxia, but this is the first time I can recall anyone connecting the hypothesis to clinical response.  Assuming that the findings can be validated in the prospective trials – and a commercial assay developed in the future – then the ability to select patients with RCC who are more likely to respond to a VEGF inhibitor such as tivozanib would be most welcome.

Would other VEGF inhibitors need to show the same effect or would a class effect be assumed?  In my book, you can’t always assume that one biomarker for one drug will always translate to others without data.  In this study a significant effect was seen for tivozanib but not sorafenib based on the hypoxia biomarker.  We don’t know what the impact is with other VEGF inhibitors such as sunitinib, pazopanib, bevacizumab and axitinib. It will be most interesting to see what happens here, as the hypoxia biomarker made be a differentiating factor for tivozanib in the future.

The upside for a VEGF inhibitor in TNBC is potentially huge, much bigger than RCC.  What would drive this element though is not only positive data but also a way to predict and select which patients are more likely to respond.  Essentially, if the data is positive, it may well redefine a subset of the disease, much in the same way that CD117 (KIT) enabled GIST patients to be differentiated from other types of soft tissue sarcomas.  When you can do that, it makes matching treatment to patients a whole lot easier for clinicians and pathologists.

AVEO presented the phase 3 clinical trial data for tivozanib in renal cancer at the American Society of Clinical Oncology Genitourinary Cancer Symposium (ASCO GU) in Orlando last week.

The TIVO-1 data (PDF download), presented by Dr Robert Motzer (New York), showed a significant improvement in progression free survival (PFS) of 11.9 vs 9.1 months (P=0.042), but not median overall survival (MOS) i.e. 28.8 vs 29.3 months (P=0.105, HR 1.25).  The lack of a significant MOS difference between the sorafenib and tivozanib treatment arms has received a lot of commentary recently, especially in light of the crossover clinical trial design.

Source:  MSKCC

Source: MSKCC

To put the clinical trial results in context, last week at ASCO GU, I interviewed Dr Motzer, (Attending Physician, Genitourinary Oncology Service, Memorial Sloan-Kettering Cancer Center, and Professor of Medicine, Weill Medical College, Cornell University) for his perspective on the data.

PSB: I am very interested in the tivozanib data from AVEO/Astellas.  What are your reactions to the results, bearing in mind that the overall survival was not significantly different from sorafenib?

Dr Motzer: The TIVO-1 study was a randomized trial of tivozanib versus sorafenib and the primary endpoint was progression free survival (PFS). Since the patients were all hoping to get tivozanib, we included in the trial design for those patients who were on the sorafenib arm, if they progressed, then we would be able to switch them over to the new promising anti-cancer drug tivozanib. What we found was that most of the patients that were progressing on sorafenib, were able to switch over and get tivozanib.

We feel that the survival being about the same on each of the two arms is maybe the result of the fact the patients on the sorafenib arm were able to receive tivozanib upon progression.

In fact, the survival was a little bit longer on the arm that got the sorafenib followed by the tivozanib, and the reason for that is that many of the patients were treated in Eastern Europe, in Russia, Ukraine, and the only access they had targeted therapies was through this study. So if they were on the sorafenib arm they were able to get both sorafenib and tivozanib, but if they were on the tivozanib arm most of them weren’t able to get any in second or third line therapy.

I think it’s important that patients get access to multiple lines of targeted therapies for kidney cancer, that is a very important thing.

PSB: Do you think, based on this data, that the drug is approvable by the FDA?

Dr Motzer: I think it should be. I think it is a very good drug, it’s effective, it has very little in the way of side effects. I think it should be an option. I think it should be approved and people should have it as an option.

Sorafenib is not one of commonly used first line agents in the United States, it has been sunitinib and now pazopanib may be the preferred agent. I would very much like to see a phase 3 trial that compares tivozanib to one of those two agents. I think that would probably provide the best information we have on how to make a choice for our patients.

PSB: What did you like about the side effect profile of this agent?

Dr Motzer: This drug is very selective for VEGF receptor, which we think is the most important target for kidney cancer response, and it has very little effects on other kinases. Many of the other drugs, they are all multi-targeted tyrosine kinases, but this one is more selective than the other ones.

Most of the side effects we see with the other drugs are from the drug hitting targets other than the VEGF receptor. So things like diarrhoea, skin sores, fatigue and so forth, that we see with the other drugs, we see very little with this one.

PSB: One of things that is noticeable in GU medicine over the last few years is the emergence of translational medicine and biomarkers, is there anything new going on in renal cancer that might help to better select patients, I think you have 8 or 9 different drugs available now, how do you choose going about which of these patients should get which of these therapies?

Dr Motzer: For the most part it is based on the evidence for efficacy and what setting the drug was studied on and it is also based on the safety profile. So if it looks like one drug is safer than another or better tolerated, then that’s often the choice to the patients.

What we don’t have is any kind of genetic profiling that can say this drug would be better for this patient and so forth. I think that is some of the work that needs to be done. There was actually one of the posters from AVEO, to try and develop some of this, like a signature for response to tivozanib. That was a poster shown here, done in a small number of patients, more kind of a pilot study. We need more of that in kidney cancer.

PSB: Do you think biomarkers will be more to the fore in renal cancer in the next couple of years?

Dr Motzer: Yes, I think the one thing is there are now a number of different medications that are similar class, they have the same mechanism of action, so I think it’s probably harder to find a biomarker to distinguish who will do better with one than another since they are related. I think the biomarkers would be important to see which patients does better with a VEGF targeted therapy like tivozanib, which one does better with an mTOR inhibitor like Torisel.

There are exciting new immunotherapies, the PD-1 antibody is very exciting in kidney cancer, for example. I think if we can identify drugs with different mechanism of action, then a biomarker might be of more use in terms of determining what is the best treatment, but when they are closely related it makes it more difficult.

The PD-1 antibody is being studied in a big global phase 3 trial by Bristol Myers Squibb compared to everolimus. There is a lot of excitement over that one. It is more of a targeted immunotherapy.

My perspective – some additional thoughts…

One of the things that got lost in the media hullabaloo when the final analysis was announced recently, was that the primary endpoint for the TIVO-1 trial was actually PFS, with median overall survival as the secondary endpoint, as Dr Motzer correctly noted.

Since tivozanib showed superior efficacy over sorafenib in the primary endpoint and a more tolerable side effect profile, then the chances of approval are higher than had both endpoints been not significant. OS is always nice, but in this disease competition is high and distinguishing between broadly equivalent agents a little harder.

In the poster, a subset analysis was presented of the North American/Western European data demonstrating MOS had not yet been reached in either arm, but 2-year survival rates were trending in tivozanib’s favour over sorafenib (75% vs. 60%). We will have to wait a little longer for the 50% point to be hit, but I thought this was encouraging. An update at ASCO in June may well be very timely.

The crossover trial design conundrum is one that other companies may well learn from – if the control arm has the potential to receive more therapy on progression that the test arm, then this will confound OS results in a global study. It’s not the first time it’s happened, but it does speak to addressing this issue more upfront in the study design. If PFS is the primary endpoint, then the issue is less of a problem but if companies need to demonstrate a clear differentiation in overall survival then a different approach might be necessary.

Several VEGF inhibitors are already approved by the FDA for the treatment of RCC (i.e. sorafenib, sunitinib, pazopanib, bevacizumab, axitinib) and in different lines of therapy, so the proof of concept for hitting the target in this disease is well established. Where they differ is that patients may well find some more tolerable than others or pricing/reimbursement may come into play as the competition heats up.

Presently, we have no valid biomarker for any of the VEGF inhibitors as a way of selecting a drug that a patient is most likely to respond to. Genentech/Roche have spent millions on biomarker research for Avastin with very little to show for it so far, proving how difficult this task really is.

Check back tomorrow for an update on tivozanib and biomarkers

Following the discussion with Dr Motzer, I followed up with Dr Murray Robinson, the Chief Scientific Officer at AVEO and will post our discussion on their fascinating research with biomarkers here on PSB. Although this research is unlikely to impact the registration trial data that is being reviewed by the FDA (the PDUFA deadline is July 28th for the TIVO-1 study), prospective inclusion of biomarkers in new studies may be very illuminating going forward.

For those of you who haven’t yet signed up to receive our conference and blog alerts, as well as information on our new cancer eReport service, you can sign up below:






Get our cancer report alerts and early bird information on oncology developments here…

We respect your email privacy

error: Content is protected !!