Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts from the ‘Uncategorized’ category

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

One way to potentially improve long term cancer statistics is earlier detection, and in high risk patients, appropriate initiation of earlier treatment, since it is well known that the survival in stage II or III breast cancer is noticeably better than that for stage IV metastatic disease.

A critical question then, is how do we improve earlier detection?

There are a number of ways to achieve this:

  1. Imaging techniques
  2. Prognostication
  3. Diagnostics
  4. Biomarkers

Historically, breast cancer has often been picked up using classic, but rather crude, imaging techniques such as mammography and ultrasound, although both have their limitations and challenges. Biopsies are also challenging and invasive, especially in early stage disease when the tumour(s) may be very small. I’m particularly interested in biomarkers because it offers a lot of untapped near-term promise. We know that as tumours begin to develop, they leave tell tall signs and signatures – how can we develop ways to detect these earlier and with greater accuracy than at present?

Source: wikipedia

I was fascinated to read a paper in PLoSONE (open access, see references below) this morning looking at circulating microRNAs (miRNA) as a potential blood based marker for early stage breast cancer detection.

miRNA were defined by Schrauder et al., (2012) as:

“MicroRNAs (miRNAs, miRs) are a class of small, non-coding RNA molecules with relevance as regulators of gene expression thereby affecting crucial processes in cancer development.”

They were first described by Lee et al., (1993) in C. elegans (open access, see references below) and have since been found to be stable in blood, making them ideal biomarker material.

In the current research, the authors set out to determine whether miRNA could discriminate early stage breast cancer (n=48) from healthy controls (n=57) using microarray analysis.

What did the research show?

The initial results appear promising:

“We found that 59 miRNAs were differentially expressed in whole blood of early stage breast cancer patients compared to healthy controls. 13 significantly up-regulated miRNAs and 46 significantly down-regulated miRNAs in our microarray panel of 1100 miRNAs.”

Two of the miRNAs (miR-202, miR-718) were subsequently validated by RT-qPCR in an independent cohort.

What do these results mean?

I thought these results were encouraging, although it should be noted that there is no doubt that blood-based miRNA-profiling is behind the improvements seen in tissue-based miRNA-profiling. The advantage though, of blood-based profiling, is that it clearly offers:

“The potential for early, non-invasive, sensitive and specific BC detection and screening.”

Of course, there is a long way to go yet, although similar early studies have been performed in other tumour types such as lung cancer (Foss et al., 2011; Boeri et al., 2011), ovarian cancer (Häuser et al., 2010) and others.

Using miRNA as a potential biomarker for early detection is not without its challenges, though. Shrauder et al., noted that Chen et al., (2008) observed that:

“Comparing serum and blood cells from the same healthy individual an almost identical miRNA profile can be found, but in cancer patients the profiles differ.”

Other studies have not shown complete congruence in the results or findings, so it may well be a while before some clarity emerges with miRNA as a potential diagnostic, most likely with improved standardisation of sample handling, protocols, detection methods and patients (stage of disease, etc).

That said, miRNA looks to be a promising but fledgling area for biomarker research in the early detection of cancer. No doubt this field will evolve further with new and more sensitive techniques.

References:

ResearchBlogging.orgSchrauder, M., Strick, R., Schulz-Wendtland, R., Strissel, P., Kahmann, L., Loehberg, C., Lux, M., Jud, S., Hartmann, A., Hein, A., Bayer, C., Bani, M., Richter, S., Adamietz, B., Wenkel, E., Rauh, C., Beckmann, M., & Fasching, P. (2012). Circulating Micro-RNAs as Potential Blood-Based Markers for Early Stage Breast Cancer Detection PLoS ONE, 7 (1) DOI: 10.1371/journal.pone.0029770

Lee, R., Feinbaum, R., & Ambros, V. (1993). The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14 Cell, 75 (5), 843-854 DOI: 10.1016/0092-8674(93)90529-Y

Foss KM, Sima C, Ugolini D, Neri M, Allen KE, & Weiss GJ (2011). miR-1254 and miR-574-5p: serum-based microRNA biomarkers for early-stage non-small cell lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer, 6 (3), 482-8 PMID: 21258252

Boeri M, Verri C, Conte D, Roz L, Modena P, Facchinetti F, Calabrò E, Croce CM, Pastorino U, & Sozzi G (2011). MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 108 (9), 3713-8 PMID: 21300873

Häusler, S., Keller, A., Chandran, P., Ziegler, K., Zipp, K., Heuer, S., Krockenberger, M., Engel, J., Hönig, A., Scheffler, M., Dietl, J., & Wischhusen, J. (2010). Whole blood-derived miRNA profiles as potential new tools for ovarian cancer screening British Journal of Cancer, 103 (5), 693-700 DOI: 10.1038/sj.bjc.6605833

3 Comments

Palm trees in downtown San Diego

Greetings from the annual American Society of Hematology (ASH) meeting in San Diego!

The palm trees and warm sunshine here were a most pleasant welcome after the bitter chill in Texas.

Having just arrived here from the San Antonio Breast Cancer Symposium, I thought it would be a nice idea to do a quick preview of some of the new and interesting data that I’m interested in at this conference and share some of the hot topics that I’ll will be following over the weekend:

  • Ponatinib in refractory CML
  • In myelofibrosis, rixuluximib and CYT-387
  • Velcade and pomalidomide in multiple myeloma
  • Obinutuzumab, PCI-32765 and alisertib in lymphomas

Let’s take a quick look at these topics in turn.

CML

The updated phase II PACE trial is expected to be presented at this meeting. Ponatinib is a potent second generation BCR-ABL inhibitor that also targets FLT3 and FGFR. The initial indication is expected to be in relapsed, refractory CML after prior treatment with a minimum of two TKIs, although many are likely to have received imatinib, dasatinib and nilotinib, so a heavily pre-treated population. Patients with advanced CML or Ph+ ALL received ponatinib in a single arm open-label trial. The other thing to note is that this agent is the only TKI so far shown to target the rare T315I mutation and these patients were included in the study.

If the results continue to hold up for durability with no additional safety signals since FLT3 and FGFR may induce off-target side effects, then I’m expecting Ariad to file ponatinib for accelerated review with the FDA, based on phase II data in relapsed and refractory CML, sometime in 2012. This is an exciting new agent and it will good for patients with CML to have an additional option in this setting.

MYELOFIBROSIS

This year has seen a lot of interest in myelofibrosis with Incyte’s ruxolitinib (Jakafi) receiving FDA approval recently. The drug was approved largely on the basis of its ability to reduce spleen size, which is one of the complications of the disease. The updated phase III COMFORT-1 data is being presented on Monday and my assumption is that we will see an improvement in overall survival with ruxolitinib.

There has been a lot of interest in YM Bioscience’s agent, CYT-387, which caused a stir at ASCO after initial data suggested that it may be able to reverse anemia associated with the condition.

Now, I’m not sure of the exact mechanism behind this phenomenon, since both compounds target JAK1 and JAK2, so the anemia response may be an artifact or a real effect. If the anemia effect is real, then I’m expecting to see the hemoglobin levels to go up rather than down, as we saw with Jakafi. The poster on Monday may well tell us more about what’s happening here and also I’m hoping to speak to some myelofibrosis thought leaders to see what their perspective is.

MYELOMA

Multiple myeloma has seen real improvements in overall survival over the last 10 years with the introduction of bortezomib (Velcade) in the upfront setting and lenalidomide (Revlimid) in the refractory and maintenance settings. Currently, a new kid on the block, Onyx’s carfilzomib, is currently being reviewed by the FDA in the refractory population, although we likely won’t know the decision until 1Q next year. If approved, it may offer physicians a new option to extend outcomes even further in advanced myeloma.

There is another agent not far behind, pomalidomide, which is a third generation immunomodulatory agent similar to lenalidomide. Celgene are presenting key data at this meeting and I’m looking forward to seeing how the data is progressing. I’m expecting this compound to show good efficacy in advanced myeloma, as it is thought to be more potent than Revlimid.

The phase III multiple myeloma study that is of great interest is the VISTA trial, which will be presented on Monday and compares the combination of Velcade, melphalan and prednisone (VMP) with melphalan and prednisone alone (MP). The five year data in treatment naive multiple myeloma will inform us which combination has superior overall survival and side effect profile and what can be expected in terms of secondary primary malignancies (SPM) with the triple versus double combination over longer term follow-up.

LYMPHOMAS AND CLL

For me, the big lymphoma story at this ASH is probably going to be GA101, now named obinutuzumab. It’s a CD20 antibody similar to, but also different from, rituximab, making it ideal for testing in NHL since the proof of concept is already established for the CD20 target.

My critical questions related to this agent’s development are:

  1. Will it overcome rituximab resistance and work in refractory patients?
  2. Will it work more effectively than rituximab earlier and prolong outcomes further?
  3. Will it have fewer side effects than rituximab?

If any of the above are true, how does obinutuzumab work differently than rituximab and does that explain any of the differences?

Not all of these questions will be answered here at this ASH meeting, but I’ll discuss these issues in more detail once the data is available.

Finally, there are a couple of other compounds in early development for lymphomas that I’m really interested in.

The first is PCI-32765 (Pharmacyclics), a bruton kinase inhibitor (BTK), while the second is Millennium and Seattle Genetics’s aurora kinase A inhibitor, alisertib. These are relatively new mechanisms of action in lymphomas and intriguing scientifically.

I’ll write more about these particular agents in depth as the data becomes available, but they’re worth watching out for over the weekend as the wires hit the news sites.

Meanwhile, you can follow the conversations at the American Society of Hematology meeting on Twitter using the official hashtag of #ASH11.

Do check back for daily updates here in the blog for the hot (and sometimes not so hot) data. I’ll also be posting a video review of the important news next week.

3 Comments

From an oncology perspective, the big drama played out in advanced high risk prostate cancer at this meeting. There were three new approvals in US last year and approval of some of them in Europe is immiment, including sanofi-aventis’s cabazitazel (Jevtana), which was approved by the EMEA at the EAU conference.

2010 and 2011 are therefore promising to be great years for increased patient and physician choice.

We took a quick look at the new data and what new therapies will likely impact the landscape for this disease in the near term, including abiraterone:

This is my first videoblog but more will be coming, including a snapshot later this week of what’s hot at the AACR meeting next weekend. Do feel free to add any comments below.

Watch this space for more!

2 Comments

Last night at the AACR keynote session at Colorectal cancer: biology to Therapy, I listened to Bert Vogelstein’s talk on the cancer genome as applied to colorectal cancer.   It was a deja vu moment of sorts, as I started my current Moleskine at AACR earlier this year with keynotes from Charles Sawyers and Bert Vogelstein. It therefore seemed most apt to finish the current volume of science notes with another talk from the same person at another AACR meeting.

The other good thing about the talk for me is that I have a few circles around squiggles from my original notes – it was good to finally figure out what the chicken scratch was trying to tell me! You can see how bad the notes were from my iPhone shot at the time; it must happen to all of us at some point – hastily scribbled notes while listening and writing in one’s lap is a recipe for disaster 🙂

Now, I’m not going to repeat Vogelstein’s talk as you can read that from the link to the April Keynote, but what interested me most about this one is that he expanded the application of what we’ve learned from the biology to patients.  This is really important, because science is ultimately about truth and how that can be applied to improving things.

Vogelstein identified three key areas that he felt had most implications for cancer patients:

  1. Cancer predisposition
  2. Identifying important biomarkers for following people with cancer
  3. Improved methods of detection

The third point is particularly relevant as it was also something Tyler Jacks talked about at the recent Xconomy meeting in Boston.  Solid tumour cancers such as colorectal cancer can take 25-30 years to develop into a metastatic carcinoma, so finding ways to identify the mass much earlier will be critical to better treatments in the future.

Future Challenges

Vogelstein expanded his ideas into some key points for future research:

  1. Improved understanding of the pathways
  2. Development of novel therapies targeting the pathways
  3. Better early detection methods

Overall, it is clear that we have come a long way over the last decade, but there is still a lot of work to do but this task will be made much easier by a larger wealth of knowledge than we started out with.

3 Comments

It’s a while since I’ve written about multiple myeloma, but after this interesting paper popped up in The Lancet Oncology, I couldn’t resist.

Many cancers occur more frequently in the elderly, a testament to the extended lifestyle many of us now enjoy compared to ancient times.  This offers many challenges compared to treating younger fitter people, not least are performance status and managing toxicities.

A standard induction treatment in untreated multiple myeloma is now the combination of bortezomib (Velcade), melphalan and prednisone (VMP), which has been shown to be superior to MP alone.  In elderly people with multiple myeloma, however, the side effects can be difficult to tolerate.

A Spanish group has considered this challenge and investigated whether a different regimen would be as effective but less intensive, thereby offering an alternative option for elderly multiple myeloma patients.

What they did was really interesting. The used bortezomib, thalidomide and prednisone (VTP) as induction therapy compared with the standard VMP followed by maintenance treatment with either bortezomib and thalidomide (VT) or bortezomib and prednisone (VP).

Here’s the treatment schedule:

The efficacy results were encouraging:

Induction:            VMP      VTP

PR                       81%     80%

CR                       28%     20%

Maintenance:         BT        BP

CR                        44%    39%

What about the side effect profile?

Overall, there was less neutropenia and thrombocytopenia in the VTP group (good news), but slightly increased cardiac events. It is also interesting to note that peripheral neuropathy (hand and foot syndrome), one of the side effects of bortezomib treatment had a relatively low incidence (less than 10%) in both groups.

Overall survival was, however, superior in the VMP group (34 months) compared with the VTP group (25 months).

As always, it is ultimately a delicate balancing act between efficacy, survival and side effects, depending of the patient’s treatment goals. The authors reasonably concluded:

“Reduced-intensity induction with a bortezomib-based regimen, followed by maintenance, is a safe and effective treatment for elderly patients with multiple myeloma.”

ResearchBlogging.org
Mateos MV, Oriol A, Martínez-López J, Gutiérrez N, Teruel AI, de Paz R, García-Laraña J, Bengoechea E, Martín A, Mediavilla JD, Palomera L, de Arriba F, González Y, Hernández JM, Sureda A, Bello JL, Bargay J, Peñalver FJ, Ribera JM, Martín-Mateos ML, García-Sanz R, Cibeira MT, Ramos ML, Vidriales MB, Paiva B, Montalbán MA, Lahuerta JJ, Bladé J, & Miguel JF (2010). Bortezomib, melphalan, and prednisone versus bortezomib, thalidomide, and prednisone as induction therapy followed by maintenance treatment with bortezomib and thalidomide versus bortezomib and prednisone in elderly patients with untreated multiple myelom The lancet oncology, 11 (10), 934-41 PMID: 20739218

1 Comment

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

via bits.blogs.nytimes.com

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

Long live the crazy ones!

(Hat tip: @mona)

Our lives are filled with the shadows of stories unwritten. Our lives are filled with waiting and hoping and putting off and stuffing the cracks with papers from other people’s lives.

If you KNEW without a doubt that your decisions were the measure of who you are – and make no doubt that they are – are you PROUD of your decisions? Are you living the story you want to live?

And what will you do with tomorrow that you haven’t done with today? What lighthouse will you point yourself towards? And what flag will you fly?

Live now. Don’t wait. And fill in some of those shadows with stories you’d want to see written about you.

Chris Brogan’s guest post on @julien’s blog today really made me stop, think and wonder.

How many of you are out there waiting for something to happen?

Think of the drug pipelines to move, the new indications to drive… Are you moving them forward purposefully or sitting back in the dog days of summer?

This is our busiest time of the year, when catching a moment to take a breather and reflect is harder, but judging by our clients some folk are out there pushing things along and trying to make a difference to the lives of people with various diseases.

Carpe Diem.

Posted via email from sally church’s posterous

"Aprepitant is the first commercially available drug of a new class of neurokinin-1–receptor antagonists for treating chemotherapy-induced nausea and vomiting. The dominant ligand for the neurokinin-1 receptor is substance P. An increase in the number of neurokinin-1 receptors on keratinocytes has been found in patients with chronic pruritus." 

 Source: NEJM

This was an interesting Letter to the Editor in the New England Journal of Medicine that I came across while looking for a different letter on JAK2 mutations and coronary ischaemia.

Essentially, the Italian authors described a brief case study where they treated two patients who were being treated with erlotinib (Roche) for non-small cell lung cancer (NSCLC) developed pruritis, ie the classic acneiform rash associated with EGFR therapy. They were non-responsive to standard systemic treatment and topical glucocorticoids.

Treatment with erlotinib was temporarily stopped while aprepitant (Merck) was given for three days, leading to "a prompt recovery" from the pruritis. Unfortunately, on resuming erlotinib with prednisone and anti-histamine prophylaxis, the pruritis returned, so the two drugs were tried concomitantly for 2 months. The severe pruritis was successfully contained (although the rash remained) and tumour responses are now being assessed.

What's interesting about this report is that normally one would not normally think of a therapy for nausea and vomiting as treatment for pruritis, but in this case, it appears to be mechanism related, ie NK1 receptors in keratinocytes found in pruritis.  The idea for the treatment came from previous correspondence to the same journal late last year regarding pruritis associated with Sézary syndrome and T-cell lymphomas.

The acneiform rash is psychologically distressing to many patients who develop it on EGFR therapy, thus prophylactic therapy that enables them to complete their anti-cancer treatment may well be a good thing, especially if they see a tumour response and, hopefully, an improvement in outcome.

In an effort to be more visible to the public, the FDA now publishes its own 'transparency blog".  This post deals with their role in regulating orphan drugs in service of the 7000+ rare diseases.

Link here.  

Fdagov_hp_logo

Posted by Bruce Lehr June 22nd 2010.

via thebigredbiotechblog.typepad.com

Saw this on Bruce Lehr's Big Red Biotech blog, which I'm a fan of.

Interesting video on the FDA blog via the link his post too.

1 Comment
error: Content is protected !!