Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts tagged ‘crizotinib’

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!

ASCO 2012 Annual Meeting ChicagoIt’s been a crazy busy time since the American Society of Clinical Oncology (ASCO) meeting earlier this month.

This year’s meeting had a lot of hidden gems in both the tumour oral and poster sessions, which will be covered in a series of blog posts.

One theme that clearly emerged was how much effort is being devoted to identifying the causes of acquired resistance to a variety of TKI single agent therapies in order to determine logical combination strategies for the clinic.

Two areas that stood out for their combined translational-clinical efforts at this years ASCO were advanced lung cancer and metastatic melanoma.

I’m delighted to announce that my first guest blogs are appearing on Chemical and Engineering News this week on these topics.  For those of you interested in advanced melanoma, check back on PSB on Thursday for the link.

The good news is that the lung cancer one has posted today. Some of the topics from ASCO 2012 covered in the post include:

Does a pan-ErbB inhibitor produce better results upfront than chemotherapy?

What new advances are there for patients with KRAS mutations?

How do we overcome ALK resistance?


Does chemotherapy produce better responses than Tarceva for EGFR wild-type in second-line NSCLC?

You can check it out here!

One of the biggest challenges facing cancer research was aptly summarised by Levi Garraway and Pasi Jänne in this month’s Cancer Discovery journal:

“All successful cancer therapies are limited by the development of drug resistance. The increase in the understanding of the molecular and biochemical bases of drug efficacy has also facilitated studies elucidating the mechanism(s) of drug resistance.”

It will therefore come as no surprise to PSB readers that resistance occurs with two drugs approved by the FDA only last year; vemurafenib (BRAFV600E melanoma) and crizotinib (ALK+ lung cancer). We’ve discussed the development of resistance in melanoma here via several potential mechanisms in the past and potential strategies for overcoming them (eg MEK inhibitors), but what about lung cancer?

Two recently published papers have shed some new light on this topic. Doebele et al., (2012) and Katayama et al., (2012) both looked at mechanisms of resistance associated with ALK-rearranged lung cancers.

What did the research show?

Both of these papers were published in March, but in separate journals.

Doebele et al., (2012) examined mechanisms of ALK resistance in EML4-ALK–positive non-small cell lung cancer (NSCLC) patients who had progressed while on crizotinib patients (n=11). The essence of their findings were as follows:

  • Four patients (36%) developed secondary mutations in the tyrosine kinase domain of ALK. Two of the patients exhibited a novel mutation in the ALK domain, encoding a G1269A amino acid substitution that confers resistance to crizotinib in vitro.
  • Two patients, including one with a resistance mutation, exhibited new onset ALK copy number gain (CNG).
  • One patient showed epidermal growth factor receptor (EGFR) mutant activity, without evidence of a persistent ALK gene rearrangement.
  • Two patients had a KRAS mutation, one of which occurred without evidence of persisting ALK gene rearrangement.
  • One patient showed the emergence of an ALK gene fusion–negative tumour with no identifiable alternate driver.
  • Two patients retained ALK positivity, with no identifiable resistance mechanism.

Meanwhile, Katayama et al., (2012) attempted to characterise acquired resistance, i.e. the adaptive resistance that occurs in response to treatment with a TKI. They also took biopsies from patients (n=18) with EML4-ALK–positive (NSCLC) patients who had progressed while on crizotinib. They found that in approximately a a quarter to a third of patients (22% to 36%) multiple mutations were found after sequencing of the ALK kinase domain exons. This resulted in amino-acid substitutions or insertions that are predicted to impair crizotinib binding. When this happens, the drug stops working and patients will relapse on therapy.

More specifically, there were:

  • Five patients (28%) had tumours with alterations in the ALK gene that were the underlying cause of the resistance.
  • There were four different somatic mutations within the ALK gene.
  • One case where the ALK gene was amplified.
  • One ALK mutation was highly resistant to all of the inhibitors examined.

In addition, they observed evidence of alternative mechanisms of resistance evolving, including activation of EGFR and KIT.

What do these results mean?

Firstly, it is striking that there are so many potential escape routes and mechanisms of adaptive resistance to crizotinib therapy.

Secondly, as Garraway and Jänne noted:

“Increased knowledge of drug resistance mechanisms will aid in the development of effective therapies for patients with cancer.”

However, while this is a true and accurate statement, I am left wondering how this might play out in clinical practice? By that, I mean how does a community medical oncologist, who sees the bulk of NSCLC patients go about incorporating this information? For now they can’t, as we are awaiting the results of numerous clinical trial readouts – hopefully there will be some at the annual ASCO meeting in June.

The sheer breadth of the heterogeneity also raises the issue of how will community doctors be able to process all this complex information and select patients for appropriate combination therapies based on numerous potential mechanisms of resistance. Biopsies aren’t always practical in these situations, but perhaps we may see the development of alternative methods of detection evolve in the future.

References:

ResearchBlogging.orgGarraway, L., & Janne, P. (2012). Circumventing Cancer Drug Resistance in the Era of Personalized Medicine Cancer Discovery, 2 (3), 214–226 DOI: 10.1158/2159–8290.CD–12–0012

Doebele, R., Pilling, A., Aisner, D., Kutateladze, T., Le, A., Weickhardt, A., Kondo, K., Linderman, D., Heasley, L., Franklin, W., Varella-Garcia, M., & Camidge, D. (2012). Mechanisms of Resistance to Crizotinib in Patients with ALK Gene Rearranged Non-Small Cell Lung Cancer Clinical Cancer Research, 18 (5), 1472–1482 DOI: 10.1158/1078–0432.CCR–11–2906

Katayama, R., Shaw, A., Khan, T., Mino-Kenudson, M., Solomon, B., Halmos, B., Jessop, N., Wain, J., Yeo, A., Benes, C., Drew, L., Saeh, J., Crosby, K., Sequist, L., Iafrate, A., & Engelman, J. (2012). Mechanisms of Acquired Crizotinib Resistance in ALK-Rearranged Lung Cancers Science Translational Medicine, 4 (120), 120–120 DOI: 10.1126/scitranslmed.3003316

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

Last week I had an enjoyable time at the AACR-EORTC-NCI Molecular Targets meeting but gippy wifi in San Francisco followed by my blog hosting and RSS feed going haywire meant that reviews of the meeting were delayed until now. There are a couple of interesting topics that emerged during the meeting that I’m going to explore in extended posts this week.

Today’s review looks at new breast cancer data from the conference. There were two things that stood out for me:

  • The role of epigenetics in advanced ER/PR+ breast cancer
  • New potential targets for inflammatory breast cancer (IBC)

Let’s take a look at these in turn.

Entinostat in second-line ER/PR+ breast cancer

The other week we discussed the data from a second generation HDAC, entinostat in lung cancer, so it was no surprise to see new data emerge in breast cancer in San Francisco as well.

Syndax reported the initial results from their phase II trial (ENCORE301) in women with hormone-sensitive breast cancer who had relapsed on an aromatase inhibitor. This is the same population recently evaluated in the BOLERO2 trial with everolimus plus exemestane at ECCO in September. In total, 49 patients were included, randomised to receive either exemestane plus entinostat (n=27) or exemestane and placebo (n= 22).

Here’s the schematic for the trial design:

schema

We know that the benefits of hormone therapy decline over time as resistance sets in. One mechanism of resistance is mTOR, and the BOLERO2 data demonstrated nicely how a logical combination of an AI with everolimus can help overcome this, leading to an improvement in progression-free survival (PFS) of 6.5 months. Hopefully, overall survival (OS) data will be available at the San Antonio Breast Cancer Symposium (SABCS) next month. Of course, as with many tumour types, there is usually more than one mode of resistance at play.

There were several key points that emerged from the epigenetics study:

  1. The ENCORE301 study is the first trial to report positive data with epigenetic therapy in breast cancer.
  2. They used a biomarker – acetylation levels – to ascertain response to therapy.
  3. Surprisingly, the clinical response to the therapy could be determined after only one or two doses.

The acetylation biomarker really intrigued me. Essentially, high levels of actylation predicted for better response with entinostat and AI therapy. The concept behind this is that HDAC inhibitors induce hyperactylation of lysines on histones as part of the mechanism of action (MOA). Thus in theory, high acetylation would potentially indicate the level of response.

What did the results actually show?

The good news is that we can see that adding entinostat to exemestane nearly doubled the PFS from 2.3 to 4.3 months, but those women with high acetlyation levels saw another doubling in the response to 8.5 months:

entinostat

Of course, this is a small exploratory study, but… the concept I think, is an excellent one, and well worth testing in a larger phase III trial.

The most obvious question that jumped to my mind after seeing the initail data was what would happen if we used a triple combination of exemestane, everolimus and entinostat or another HDAC in this relapsed population?

I don’t know the answer, but would love to see a phase II study emerge to get a quick readout on the possibilities. Many of you will recall that:

a) The Wyeth mTOR trial with temsirolimus in several thousand women with breast cancer produced a resoundingly negative result, but that that was in the front-line setting and mTOR is activated over time, causing resistance.

b) Merck’s HDAC inhibitor (vorinostat, SAHA) was evaluated in several breast cancer trials and none of those produced a positive result as far as I recall. That begs the question – was it the trial design or the drug – not all HDACs may be equal.

The good news here is that there is both a positive result and also a biomarker of response. Those suggest that it would be worth testing further in the relapsed setting both as a doublet in a large phase III study and in triple combination with everolimus in a smaller phase II trial.

Overall, I was very impressed with these results and Syndax should be congratulated for an excellent study design and also developing a useful biomarker. Neither are easy to do well.

Is ALK a new target in inflammatory breast cancer (IBC)?

This one caught me completely by surprise. IBC is a rare, but rather nasty, form of breast cancer that is often diagnosed late (in stage IIIb/IV). It presents with red, inflamed and thickened skin, rather than with a tumour, like this:

IBC

Sadly, we still have a lot of progress to make in understanding the aetiology of this disease, which often shows an accelerated path to metastasis, although we don’t know why. There aren’t that many new therapies or clinical trials in this area either as a rsult of the paucity of knowledge around the biology.

Dr Fredika Robertson (MD Anderson Cancer Center) presented the initial results of some translational research in a small number (n=12) of women with IBC.

She suggested that the early evidence is that the ALK translocation may be a transforming oncogene in breast cancer.

What did they find?

As a result of earlier work from Perez-Pinera et al., (2007) showing ALK gene expression in several types of breast cancer, they decided to look at this more closely in both pre-clinical animal models and also IBC patients.

These are the initial findings in women with IBC:

ALK

Note that they found an incidence of 75% for the ALK translocation in the dozen patients tested. I personally would be leery of extrapolating the results from such a small sample size to the broader population, but it certainly would be worth investigating further.

There are several questions that come to mind:

  1. Is the effect real or not? See Krishnan et al’s (2009) paper on intravascular ALK-Positive Anaplastic Large-Cell Lymphoma mimicking inflammatory breast carcinoma (reference below).
  2. Is the ALK translocation a key driver of aberrant activity?
  3. If yes, would an ALK inhibitor be effective or not?

In order to answer the last question, there is a multi-centre phase I trial with LDK378 (Novartis) now enrolling patients with ALK+ positive advanced cancer to find out the answer. In addition, Dr Robertson mentioned a single centre trial with crizotinib in ALK+ breast cancer, although I couldn’t find it in the clinical trials database.

Conclusions:

Overall, it was good to see some new progress being made in both translational research and also in the clinic, albeit the results are still early, but rather encouraging I think.

These two concepts, ie epigenetic therapy in ER/PR+ breast cancer and ALK translocations in IBC, will be worth following over the next couple of years to see whether they progress our knowledge and eventually more effective and targeted treatments of different subsets.

In the meantime, a further update of exciting new developments in breast cancer will be posted on this blog next month from the San Antonio Breast Cancer Symposium (SABCS).

References:

ResearchBlogging.org Perez-Pinera, P., Garcia-Suarez, O., Menendez-Rodriguez, P., Mortimer, J., Chang, Y., Astudillo, A., & Deuel, T. (2007). The receptor protein tyrosine phosphatase (RPTP)β/ζ is expressed in different subtypes of human breast cancer Biochemical and Biophysical Research Communications, 362 (1), 5-10 DOI: 10.1016/j.bbrc.2007.06.050

Krishnan, C., Moline, S., Anders, K., & Warnke, R. (2009). Intravascular ALK-Positive Anaplastic Large-Cell Lymphoma Mimicking Inflammatory Breast Carcinoma Journal of Clinical Oncology, 27 (15), 2563-2565 DOI: 10.1200/JCO.2008.20.3984

6 Comments

“Ipilimumab is not recommended for the treatment of advanced (unresectable or metastatic) malignant melanoma in people who have received prior therapy.

The Committee was satisfied that ipilimumab meets the criteria for being a life-extending, end-of-life treatment and that the trial evidence presented for this consideration was robust.

The Committee acknowledged that few advances had been made in the treatment of advanced melanoma in recent years and ipilimumab could be considered a significant innovation for a disease with a high unmet clinical need.

Despite the combined value of these factors the Committee considered that the magnitude of additional weight that would need to be assigned to the QALY gains for people with advanced (unresectable or metastatic) melanoma would be too great for ipilimumab to be considered a cost-effective use of NHS resources.”

NHS NICE Guidance

In other words, it’s too expensive and the NHS doesn’t want to pay the £80K ($120K) sticker price. This news is no great surprise given the cost-benefit ratio when considering that there is no way to tell who might benefit most from treatment upfront.

The is, however, a huge difference between hope and false hope, as NPR Shots astutely noted when discussing Avastin in breast cancer earlier this week and in some ways that sentiment applies here too. By this I mean it would be much more compelling to both patients and NICE if an oncologist could talk about a new therapy in specific and useful terms.

Examples of doctor-patient conversations about treatment in the near future might look more like this….

Either:

“You have an 70-80% chance of responding to this therapy because you have X (mutation, translocation, biomarker etc), where this drug has been shown to be highly effective and extends life by over 2 years in many of our advanced patients with this disease to date.”

Or:

“This drug may do more harm than good in your case, as it has been shown to effectively target X (mutation, translocation, biomarker etc), which you do not have, and therefore, are unlikely to respond. I believe it would be better to consider these alternatives in your situation… “

We all know about heterogeneity – it’s the very underpining of what makes a cancer survive despite our best efforts to tame it until we can subset into more homogenous and predictable groups.  This means that offering a broad therapy to all patients with a given advanced cancer without any idea of its predictive value is fast becoming a misnomer in today’s world of emerging targeted therapies.  Now, manufacturers (marketers even) might think it’s better not to ‘limit’ their market opportunity, but the reality is many healthcare systems are looking at ways to limit treatments to where it’s needed most, not only for cost reasons, but also to direct resources where they are more likely to work. The current model is not sustainable in the long run.

Of course, if a predictive biomarker was available to determine which patients are more likely to respond to ipilimumab, then the QALY calculation would be considerably different, and possibility even within the realms of the current guidelines.

That’s a whole different ballgame, but hopefully one that will begin to emerge as we have seen with new targeted therapies such as vemurafenib (Zelboraf) in BRAF V600E malignant melanoma, crizotinib (Xalkori) in ALK-positive advanced lung cancers and everolimus (Afinitor) in combination with exemestane in ER/PR+ HER2- breast cancers that have relapsed after initial aromatase inhibitor therapy.

It will be interesting to see how NICE handles all of those situations in the future, since they are all targeted agents showing a significant impact on a patients ability to live longer,with a more precise, and therefore, limited patient definition.  As a Brit and a scientist, I may have reasonable expectations that NICE will make a rational and logical decision in the face of limited resources, but this is also tinted with a large dose of healthy scepticism after the trastuzumab (Herceptin) debacle in HER2-positive breast cancers that lead to the utterly ridiculous and unfair post code lottery in the UK.

We are not talking absolute costs here, but the relative costs of seeing real efficacy benefits of six months or more in those patients most likely to respond, while at the same time giving an offering that truly extends life in a meaningful fashion without exposing too many to the toxic side effects of a given treatment. Dealing with cancer is tough enough without being treated with a regimen that had absolutely no hope of helping people live longer and feel better.

Pfizer’s Crizotinib (Xalkori) approved in ALK-positive lung cancer!

The FDA just announced that they have approved Pfizer’s crizotinib (Xalkori):

The U.S. Food and Drug Administration today approved Xalkori (crizotinib) to treat certain patients with late-stage (locally advanced or metastatic), non-small cell lung cancers (NSCLC) who express the abnormal anaplastic lymphoma kinase (ALK) gene.

Xalkori is being approved with a companion diagnostic test that will help determine if a patient has the abnormal ALK gene, a first-of-a-kind genetic test called the Vysis ALK Break Apart FISH Probe Kit. It is the second such targeted therapy approved by the FDA this year.

Source FDA

This is wonderful news for those unfortunately affected by this debilitating disease and those yet to be diagnosed with the aberration who will be able to be treated with a new highly specific and targeted drug.

ALK aberrations typically occur in the order of 4-7% of NSCLC patients, depending on sources. No doubt the companion FISH diagnostic test from Abbott will make it easier to screen and identity patients. In turn this will help determine which patients with lung cancer are eligible for treatment.

Pfizer began the rolling NDA submission in January and completed it in May, giving a PDUFA date around November 17th. This rapid approval in approx. three months continues the 2-3 month trend seen with cabazitaxel (Jevtana) and abiraterone (Zytiga) in castration-resistant prostate cancer (CRPC), and vemurafenib (Zelboraf) in metastatic melanoma.

The Xalkori story has been nothing short of amazing and represents another major advance for targeted therapy in a clearly identified subset of patients. There are several patient stories that I’ve come across on the internet, most are heart warming – take a look at this snippet I have curated from ‘feel good’ anecdotes from a caregiver this month alone:

View “Does crizotinib work in ALK+ lung cancer?” on Storify

It’s amazing to follow their story of courage and grace under pressure; it is also very hard to have a bad hair day when the very fragility of human life stares at you in the face. It could be any of us under 50, even non-smokers.

The response rates to crizotinib have been incredible, as witnessed by Dr Jack West’s story about one of his patients at Swedish:

{Update 1: Dr West tells me that the young gentleman he referred to in his TED story has now been on crizotinib 2+ years and is doing well enough to coach soccer!}

In the final PI, the overall response rates for Xalkori in two single arm studies (n=136 and 119) in patients who had mostly received prior systemic therapy. They differed in that:

“In Study A, ALK-positive NSCLC was identified using the Vysis ALK Break-Apart FISH Probe Kit. In Study B, ALK-positive NSCLC was identified using a number of local clinical trial assays.”

The ORR was 50% and 61% for each respectively. This is pretty impressive, in my opinion. According to the PI, the adverse event profile is quite tolerable:

“The most common adverse reactions (≥25%) across both studies were vision disorder, nausea, diarrhea, vomiting, edema, and constipation.
Grade 3-4 adverse reactions in at least 4% of patients in both studies included ALT increased and neutropenia.”

These are fairly normal and commonplace for cancer therapy, although there are potential vision disturbances that may need to be watched (from the PI):

“Vision disorders including visual impairment, photopsia, vision blurred, vitreous floaters, photophobia, and diplopia were reported in 159 (62%) patients in clinical trials.
These events generally started within two weeks of drug administration.
Ophthalmological evaluation should be considered, particularly if patients experience photopsia or experience new or increased vitreous floaters.
Severe or worsening vitreous floaters and/or photopsia could also be signs of a retinal hole or pending retinal detachment.”

The good news is that Xalkori is now available – according to the Pfizer press release:

XALKORI is available immediately through a number of specialty pharmacies.
Patients prescribed XALKORI can call 1-877-744-5675 for assistance accessing the medication.

For more information about the FDA-approved ALK test, call (855) TEST-ALK (837-8255).

The big question many will be asking, though, is what’s the price?

Answer: Price: $9,600/month, putting it in line with similar pricing to Roche’s Zelboraf in metastatic melanoma.

{Update 2: Abbott has also received approved for the ALK test although no information on the cost of the test was provided}.

 

13 Comments

“The problem at the moment is that it takes $1bn [£600m] to get a drug to market and 15 years or more. That is the justification for the pharmaceutical industry charging high prices.

If on the other hand by the time you get to phase 2 you know exactly which patients it is going to work on, you only put those patients through and instead of 10% you get an 80% response rate.

You get a licence on the basis of the data and don’t have to go to phase 3 (a trial involving thousands of people). That saves vast sums of money and years of development. What that does to the business model is it means you can justify charging lower prices because it cost a lot less in the first place.

If we get this right, it changes the entire dynamics of the business model of the pharmaceutical industry.”

Source Harpal Kumar, the chief executive of Cancer Research UK (CRUK) via The Guardian

A UK friend kindly sent me this article today and provocatively asked me what I thought. Hmmm, a very interesting, meaty and relevant topic indeed.  Here goes…

Will this change the way we do business in cancer research?

The theory behind this statement by CRUK is that if we develop more targeted drugs to fewer patients and generate higher response rates e.g. 70-80% in a specific biologic subset, instead of say, 10% in a broader population, then the costs of development will come down and thus the treatment cost of the disease will ultimately lower.

Not so fast!

The reality might actually be different, and here’s why:

  1. For this to happen you need more translational research, biomarkers and companion diagnostics.
  2. The cost of researching and developing the targets is quite high.
  3. While clinical development costs might be lower with fewer large scale trials, the costs of iterative phase II trials will go up and the available pool of patients for commercialization is now much lower e.g. 5% of patients with the ALK translocation in lung cancer not 100% of all available relapsed patients.
  4. In order to maintain revenues, it is basic economics 101 that smaller patient niches will equal higher costs.

If you are not convinced of the last point, take a look at the costs of treating rare diseases or small subsets of patients.

Some good examples exist in the hematology space include:

  1. Alexion’s Soliris (eculizumab),which is approved for the treatment of patients with paroxysmal nocturnal hemoglobinuria (PNH). This is a rare hematologic disorder that affects approx. 8,000 to 10,000 people in North America and Europe. The cost is something like $400K per annum.
  2. Genzyme’s Fabrazyme (agalsidase beta) for the treatment of Fabry Disease, another rare hematologic condition, this time an inherited metabolic defect that affects 1 in every 40-50,000 people in the US. Fabrazyme lowers the amount of a substance called globotriaosylceramide (GL-3), which builds up in cells lining the blood vessels of the kidney and certain other cells. It’s very effective, but certainly not inexpensive at around $160K per annum.

What is the likely impact of the changing research paradigm?

Both of the above patient pool sizes are not out of the realm of reality for a comparison with oncology.

In the old model, clinical trials tended to involve more allcomer trials, i.e. patients with a particular tumor type (e.g. non-small cell lung cancer), stage of disease (metastatic) and line of therapy (frontline, relapsed or refractory).

In the new world order, things are changing in clinical trials already:

  1. Roche’s Zelboraf (vemurafenib) was recently approved in metastatic melanoma in patients with the BRAF V600E mutation, reducing the available pool who might respond by 50%. It was launched last week with a price tag of around $56.4K for an average of 6 months treatment.
  2. Crizotinib (Xalkori) is being evaluated in patients with NSCLC who have the ALK translocation and have failed prior therapy. That’s a tiny subset of patients. Patients with this aberration make up maybe 4-7% of the total NSCLC pool. Imagine how small the target population will be for other ALK inhibitors in crizotinib refractory disease?!
  3. The cost of funding and finding biomarkers that predict response is a huge undertaking.  Genentech have no doubt spent many millions looking for a predictive biomarker for Avastin, so far to little avail.

Of course, there are plenty of other exciting targets with small subsets being evaluated in the clinic, but there are several factors to consider:

  1. Small subsets = fewer patients = higher cost.
  2. Will combination strategies be affected by the cumulative costs that will inevitably result? e.g. Yervoy + Zelboraf in metastatic melanoma potential treatment cost = $170-180K if the studies are successful in showing that survival is improved.
  3. Since Dendreon’s Provenge ($93K) was recently given the green light by the CMS, the costs of new targeted entrants is creeping up over the $100K watershed marknot down, viz Yervoy ($120K) and Adcetris (~108K), for example.

In conclusion…

I admire the chutzpah of CRUK, but disagree with some of their conclusions, which I think are rather naive.

Today, I will go on record here and declare that I believe specialised treatment based on the underlying biology will ultimately cost more, not less, in the long run in terms of research and development, diagnostics/biomarkers and treatment costs of ever smaller subsets.

However, I have no doubt we will ultimately see better results clinically with this more scientific approach but this will come at a cost.  While that’s great news for patients and caregivers, it is not so great for the payers, Government or investors, because higher risks and R&D costs will inevitably equate to more failures and this drives higher costs in a spiral fashion.  Ultimately, those costs will trickle down to all of us in the form of higher co-pays and more expensive medical plans to cover the payers margins.  Success has to be paid for somewhere down the line.

And the constant refrain from everyone in Pharma of “let’s do more with less” will increase.

It’s a vicious cycle of unsustainability, with no end in sight.

9 Comments

This morning, Seattle Genetics announced that the expected fast track approval from the FDA has been forthcoming for brentuximab vedotin (Adcetris) following the recent unanimous ODAC voting in both refractory Hodgkin Lymphoma (HL) and Anaplastic Large Cell Lymphoma (ALCL).  Clearly, the company and the agency have come to agreement on the confirmatory trials as part of the condition of accelerated review.  The final prescribing information (PI) can be found on the Adcetris website.

For those of you looking for more information on Adcetris, please check out the related posts section below for previous reports on this novel ADC therapy.

I missed the conference call this morning announcing the pricing details, but Luke Timmerman from Xconomy had a nice summary:

“The company set the price at $13,500 per dose, given intravenously every three weeks. If patients get eight infusions on average, consistent with clinical trial experience, then it will cost $108,000 per patient.”

Based on the data from the clinical trials, most patients will probably need 7-9 cycles, so this would make the overall course in the $94,500 – $121,500 per treatment range, with $108K being the average based on 8 cycles.  The overall treatment cost will therefore typically be less than the $120K cost of treatment for BMS’s ipilimumab (Yervoy) in metastatic melanoma.

Ever since Dendreon announced the $93K course of treatment for Provenge in asymptomatic castrate resistant prostate cancer (CRPC), there has been a noticeable trend upwards in the cost of treating advanced cancers.  We probably spend more treating the last 6 months of a cancer patients life than any of the other stages combined with incremental rather than dramatic improvements.  In the long run, this is likely to be unsustainable, but for now companies will continue to charge what they think the market will bear.

The good news is that Adcetris offers excellent clinical proof of concept for antibody drug conjugate (ADC) technology and has the distinction of being the first such agent approved, beating Roche’s T-DM1 to market, which has been filed with the FDA for the treatment of HER-2 breast cancer.

In the meantime, we also have several other novel therapies awaiting final review by the FDA.  Aside from T-DM1, Pfizer filed crizotinib for ALK-positive lung cancer and now has a brand name, Xalkori, which I thought sounded rather Vulcan :).  Should these two agents also receive FDA approval in the very near future, 2011 will likely be a bumper year for new cancer drug approvals.

 

2 Comments

Love this video from TEDxOverlake, where Dr Jack West (Swedish) describes what he is doing with his excellent forum and site, the Global Resource for Advancing Cancer Education (GRACE).  Currently, the main focus is on his specialty, Lung Cancer, but more tumour types are planned in the near future.

Jack talks about how physician led sites can actively and effectively reach out in a many-to-many fashion to improve education and learning, rather than in the traditional one-to-one fashion seen in a consultation.

He uses a great example of how a young patient with lung cancer was empowered to seek out better care for his condition and ended up in the crizotinib trial for ALK+ non-small cell lung cancer…

Check it out – it’s a compelling and very powerful story:

If you can’t see the video in your email subscription, you can find it here.

 

Disclosure: I’m an unpaid member of the GRACE Advisory Board (so of course, I will be biased 😉

 

error: Content is protected !!