Pharma Strategy Blog

Commentary on Pharma & Biotech Oncology / Hematology New Product Development

Posts by MaverickNY

Following this morning’s blog post, I’ve been deluged with emails wanting to know more about the basics of miRNA and it’s role in cancer.  It’s a very complex topic, as Walter Jessen, a cancer researcher pointed out on Twitter in a discussion with Angela Alexander, a Ph.D student at MD Anderson and myself about miRNA:

“Complicated stuff, esp when expression of mirs doesn’t jive with expression of genes that reg them or that they reg.”

Where reg is ‘regulate’ since Twitter only allows 140 chars or less for communicating ideas between people 🙂  Still, this was exactly the problem I was experiencing when researching the topic recently.

Walter kindly offered the link to a primer he wrote on his excellent blog, Highlight Health, which has some great cancer resources and information.  Do check it out.

For those of you already familiar with the technical research in this area, there is a nice blog site specialising in miRNA that is well worth checking out for resources and information.

Memorial Sloan Kettering Cancer Centre in Manhattan have a useful online search tool, which looks at “Predicted microRNA targets & target downregulation scores. Experimentally observed expression patterns.” Be warned, you can become distractible and lose yourself for a hour or two playing with this one 🙂

The University of Southampton in the UK also have a Junk RNA blog site collating some information as well.

Wikipedia has some notes on the topic and some useful diagrams like this one:

Source: Wikipedia

Please feel free to add any other useful resources for others to browse in the comments section below.

After last weeks big picture, strategic look at the state of cancer research, I thought it would be a nice idea to go the other extreme and look at some promising work that is happening at the cellular level.

microRNA is becoming an increasingly hot topic in oncology.   Technically, they are are post-transcriptional regulators that bind to complementary sequences in messenger RNA.  I tend to think of them more simply as gene regulators otherwise my brain starts to fry round the edges a bit.

Recently, He et al., (2005) demonstrated that when mice were altered to produce more Myc protein, miRNA was involved in the development of cancer.  Mutated Myc has been implicated in several cancers.  They engineered the mice to produce a surplus of types of miRNA found in lymphoma, ie mir-17-92 polycistron, and noticed that enforced expression of the mir-17-92 cluster acted with c-myc expression to accelerate tumour development.  This miRNA cluster may therefore act as an oncogene and represent a valid therapeutic target in lymphoma.

Several miRNA’s have now been implicated in cancer.  You will often see them numbered, eg miR-7, for ease of recognition.  Last week, I came across another interesting article on miRNA and lung cancer.  We know that EGFR plays a key role in the growth of epithelial cells, and faults in the network via over expression or mutation can lead to signalling overdrive and eventually lung cancer.  One of the big questions is how do miRNAs mediate EGFR signalling to modulate tumorigenesis?

Chou et al., (2010) took a different approach from He et al.  They looked at the interplay between EGFR, miRNA and Myc in lung cancer:

“To search for miRNAs regulated by EGFR, we performed miRNA microarray analysis of EGFR-silenced CL1-5 cells, a lung cancer cell line that overexpresses EGFR.  Strikingly, we found that miR-7 is highly induced by EGFR in lung cancer.  In the present study, we show for the first time that deregulated EGFR signaling induces miR-7, which in turn suppresses ERF and plays an important role in the oncogenesis of lung cancer cells.”

miRNA in the form of miR-7 is interesting, because it was previously found to regulate cell growth and apoptosis of cervical cancer and neuroblastoma cells.   In this study, miR-7 was found to be overexpressed in human lung cancers and negatively correlated with disease free survival, echoing an important role of miR-7 in cancer progression.

One conundrum that can cause confusion is the finding that EGFR overexpression or amplification occurs more frequently in squamous cell carcinoma than in adenocarcinoma, but EGFR mutation occurs mostly in adenocarcinoma see Dacic et al., (2006).  Chou et al., found that the level of miR-7 correlated with EGFR expression in squamous cell carcinoma and EGFR mutated adenocarcinoma, supporting the idea that deregulated EGFR signaling induces miR-7 expression.

In addition, miR-7 has been shown to suppress EGFR expression and function as a tumour suppressor in glioblastoma, but several reports have also shown that miR-7 expression correlates with poorer prognosis in patients with breast cancer and urothelial carcinoma.  It is not impossible that miR-7 may therefore play different roles in different cancer types.

Another observation that caught my eye was:

“We have found that blocking the EGFR-mediated phosphoinositide 3-kinase (PI3K)/AKT pathway with LY294002 inhibitor also attenuates miR-7 expression, suggesting that the PI3K/AKT pathway participates in miR-7 regulation.”

There has been a recent flurry of interest in the roles of PI3K, AKT and MEK inhibition, especially in lung cancer, behind the research scenes.  There are now more PI3K inhibitors in development than I can remember, but the key thing will be understanding the biology and translating it to well designed studies, otherwise we will be back to the sad old flawed model of combine it with standard chemo and then suck it and see.  That sledgehammer to crush a grape approach is unlikely to work here, so we may see quite a few failures before someone cracks the code and designs some really smart studies.

I do think this is a very interesting pathway (as you can see from the many posts on the topic in the archives such as this one) and one that we may well see more results from the clinic in the future, once the research points to a clearer path for more logical combinations.  Being first into the clinic doesn’t necessarily mean it’s the right approach.  Sometimes, sitting back and working out the best approach first may lead to better results in the long run.

Finally, in the discussion, Chou et al., noted:

“In this study, we have shown that EGFR stimulated miR-7 expression via the Ras/ERK/Myc pathway to promote cell proliferation, anchorage-independent cell growth, and tumor formation of lung cancer cells.”

It will be fascinating to see if miR-7 emerges as a new prognostic biomarker and helps to provide a new valid therapeutic target in lung cancer.  If you are interested in this field of research, do check out the links to the references below.

References:

ResearchBlogging.org He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ, & Hammond SM (2005). A microRNA polycistron as a potential human oncogene. Nature, 435 (7043), 828-33 PMID: 15944707

Dacic S, Flanagan M, Cieply K, Ramalingam S, Luketich J, Belani C, & Yousem SA (2006). Significance of EGFR protein expression and gene amplification in non-small cell lung carcinoma. American Journal of Clinical Pathology, 125 (6), 860-5 PMID: 16690485

Chou, Y., Lin, H., Lien, Y., Wang, Y., Hong, C., Kao, Y., Lin, S., Chang, Y., Lin, S., Chen, S., Chen, H., Yeh, S., & Wu, C. (2010). EGFR Promotes Lung Tumorigenesis by Activating miR-7 through a Ras/ERK/Myc Pathway That Targets the Ets2 Transcriptional Repressor ERF Cancer Research, 70 (21), 8822-8831 DOI: 10.1158/0008-5472.CAN-10-0638

1 Comment

The title of this post today is inspired by one of my bioinformatician science buddies on Friendfeed, Neil Saunders, who has a great blog that’s worth checking out.

Here’s a wonderful simplified picture of many of the pathways thought to be involved with different types of cancer and was shown by Dr Wafik El Deiry of Penn at the recent AACR meeting on colorectal cancer in Philly:

Source: AbCam (pdf download)

Imagine all those pathways that are overexpressed in any given cancer, some may well be mutated, most will be passengers, a very few will be actual drivers.  Now imagine that all of them are lit up like a Christmas tree.

Not so easy to see the wood from the trees now, is it?

Think about drug development… how many people rush off and pick off one target and take a targeted therapy, either a monoclonal antibody or kinase inhibitor, combine it with standard chemotherapy and think it possibly might work, let’s suck it and see.

Ummm, no.

What are the chances of such a random unscientific approach actually working?  Pretty low.  Then we sit back and realise that the old models (animal, clinical etc) aren’t working any more and such an approach is no longer sustainable.  The drain on resources, whether time, money or people is too high.  The phase II/III attrition rate with that throw the mud at the wall approach is horrendous in oncology.

There is another way.

The smart researchers and companies are now using more modern, highly evolved animal models, doing more extensive preclinical research and thinking differently using a more holistic systems biology approach.  They’re trying to figure out what the logical drivers are, which might suggest some logical combinations (think two unapproved targeted agents with an approved one perhaps) based on the constitutively activated  or mutated targets, cross-talk, feedback loops and going into phase I research later with a more solid rationale.

The winners in this will be the companies with the most useful and broad pipeline who can mix and match more easily in this strategic pathway rather than tumour approach, with their own compounds than someone else’s.  Collaborations on the pharma side are still relatively few and far between.  They are often also a nightmare to manage, no matter what the original intentions were.

Change is already happening judging by the many enlightening conversations I’ve had this year with academic and pharma researchers, clinicians and commercial clients alike.  This is great news and it’s driven by a greater understanding of basic research, better animal models, a panopoly of potential druggable targets and a broad, deep pipeline across the oncology companies as a whole.  We just need to start putting the jigsaw together now and maybe we’ll see a difference in outcomes in the not too distant future.

The future may not be so far off as we think.  The future is now.

10 Comments

This is meant as a light hearted fun post, hat tip to Coturnix (Bora Zivkovic).

For those of you unaware of who the identity of the Pharma Strategy Blog writer, MaverickNY, is – well, apparently according to this analysis, I’m likely to be male, 66-100 yo and a happy academic.  LOL I must have been to too many science conferences lately 🙂

You can check out the origins of your own blog or website here.

5 Comments

A monk told Joshu, “I have just entered the monastery. Please teach me.”

Joshu asked, “Have you eaten your rice porridge?

The monk replied, “I have eaten.”

Joshu said, “Then you had better wash your bowl.”

At that moment the monk was enlightened.

Source: NoZen

It’s not a well known fact that I love minimalism as a concept, although I rarely achieve that nirvana, and have enjoyed reading many books on Zen.

The topic of mindfulness and cancer was something that came up in conversations at ESMO recently with Dr Steven Tucker.  He made some interesting points about mindfulness for health, as well as disease, and that’s something I have been thinking about more since then.

Yesterday, though, I went out for an early quiet lunch at a local Japanese restaurant thinking some quiet reflection would be a pleasant antidote to a busy couple of weeks on the work front.

You might think that the concept of peace, quiet and focused mindfulness would fit well with the cuisine.

Instead, after sitting down at the simple black tables, someone cranked up the sound on the TV in the corner of the room.  Service was fussy and overly intrusive.  As I was finishing, as soon as I barely put my chopsticks down in the Bento box, a waitress rushed over to snatch it up.  There was little sense of the peace and relaxation that I craved for while out of the office.

Later, while sipping some Jasmine tea in a little Chinese tea cup in the quiet of the office, some more reflective thoughts made me wonder – how many people actually stop and reflect in the hurly burly of life as it passes by at a frenetic pace?  How much does inner stress and outwardly fast paced lifestyles contribute to the origins or acceleration of cancer?

Perhaps we all need more quiet, focused mindfulness in our lives.  This is something I plan to work on more myself.

What about you?

18 Comments

For those of you interested in life science VCs, M&A and IPO’s, the latest OnBioVC report is now available for download here (pdf).

Inevitably, in the current economic climate, the financial trend is a downwards one, as evidenced by the findings:

“The 3Q10 OnBioVC Trend Analysis tracked, in aggregate, 70 biopharma, diagnostic, device, medical-IT and biofuel venture financings totaling $1,420.0M. Compared to 3Q09 OnBioVC data, this investment activity represents a decrease in the number of quarter-over-quarter financings by 25 (95 v. 70) and a decrease in quarter-over-quarter invested capital of $242.9M ($1,420.0M v. $1,662.9M).”

What was interesting to me though, was that oncology still looks a robust area compared to most sectors:

“For 3Q10 the oncology sector continued to flex its muscles, accounting for one in five deals closed in the quarters ninety days, the 14 financings accounted for $308.1M and interestingly approximately 22% of capital invested.”

The largest oncology deal involved Immatics Biotechnologies who closed a $71M (USD) Series C financing.  This interesting German based company is developing cancer vaccines in renal, colorectal and glioblastoma.

Aside from Immatics, other companies developing cancer therapeutics who raised money in the last quarter include some familiar and not-so familiar names:

  1. Alethia Biotherapeutics
  2. Anchor Therapeutics
  3. Calistoga
  4. Celator
  5. Cylene
  6. Dicerna
  7. Eddingpharm
  8. Geming X
  9. Jennerex
  10. TetraLogic
  11. Tigris

Calistoga Pharma are developing PI3 kinase inhibtors, including CAL-101, which I have blogged about at ASCO earlier this year and on the PI3K class in the past. They are currently evaluating the drugs in cancers such as B-Cell Lymphoma and Leukemia, as well as immunology indications.

Dicerna Therapeutics are involved with developing new therapeutics based on siRNA and some of you may remember them from my recent report of the Xconomy ‘War on Cancer’ meeting in Boston.

I’m not going to spoil the findings of the OnBioVC report, but check it out as there is a wealth of information contained therein and it’s worth a quick read to see the general trends in this area.  There is also a lot of useful information on their website.

1 Comment

This week heralds a busy one with the upcoming annual NY Chemotherapy Foundation (Greenspan) meeting that begins tomorrow at the Marriott Marquis in Times Square with the Pediatric session in the afternoon and finishes on Saturday with the Nursing session.

I’ll be at the Greenspan event from Weds to Fri, please do say hello if you see me around.

In the meantime, I’ve been pleasantly surprised at the number of signups to the conference newsletter (see the sign up widget in the right hand margin —>).   The first one, from AACR on colon cancer, goes out this on Wednesday week, please sign up before then if you would like to receive it.

More newsletters will follow soon on the Greenspan Meeting, ASH and SABCS.

I’ve just added a new meeting to my January conference schedule and will be attending the 6th International Society of Gastroenterological Carcinogenesis in Houston, Texas. It’s hosted by MD Anderson and you see the program here.  Many thanks to Drs Ray DuBois and Anas Younes for alerting me to the event.  There are some interesting speakers and topics so it looks like being a good meeting.

You can see my active schedule in the Conference Schedule tab at the top of the site. More events will be added for 2011 as dates firm up.

2 Comments

This morning I was delighted to see that one of my favourite medical doctors on Twitter, Dr Anas Younes from MD Anderson, has published a paper in the New England Journal of Medicine on a clinical trial of a promising new agent in development for a particular type of lymphoma.

Dr Younes is very active in social media on Twitter and Facebook and has garnered quite a following of lymphoma patients interested in learning about new treatments for the disease.   This also means that patients and caregivers following him are able to find out about new clinical trials as they open up.  MD Anderson Cancer Center probably has access to more clinical trials across all tumour types than any other cancer center, offering lots of options for cancer patients to receive novel therapies that may help their condition.   Another benefit of a physician being involved with social media is that awareness of the trials will reach more people this way and hence probably accrue faster.

What’s new in Hodgkins Lymphoma?

Before we discuss the NEJM paper, the other side of social media is that MD Anderson also use it to communicate the results of their trials on the Institution website and Dr Younes also has a nice video for patients, explaining how the drug works, about the trial and the results that they found.

Now, this was a phase I trial so normally we wouldn’t expect to see too much from an efficacy standpoint, as the main goal of these studies is to assess the range of toxicities and determine the maximum tolerated dose (MTD) for phase II trials, which look at the efficacy signal in more detail.

That said, what Dr Younes and his colleagues found was quite impressive responses in a disease that has not seen much in the way of new treatments for more than a decade.

It should be noted that the agent targets CD30 antigen that is expressed on Hodgkin Lymphoma and anaplastic large-cell lymphoma (ALCL) cells.  Previous attempts to target the CD30 antigen with monoclonal-based therapies have shown minimal activity.  What’s different about this new agent is that it is an antibody-drug conjugate (ADC), with an anti-CD30 monoclonal antibody linked to monomethyl auristatin E (MMAE), an anti-cancer agent.

The drug they were evaluating, brentuximab vedotin (SGN-35), from a partnership between Seattle Genetics and Millennium, elicited complete responses (CR) or partial responses (PR) in 38% of the patients with Hodgkin Lymphoma (HL).  In the NEJM article it boldly stated that:

“The median duration of response was at least 9.7 months. Tumor regression was observed in 36 of 42 patients who could be evaluated (86%).”

Looking at the data in the article, what was amazing was that there were 17 objective responses (38%), 11 of which (25%) were complete remissions, which essentially means disappearance of all evidence of the disease.  In addition, CT scans showed that 36 of 42 (86%) of evaluable patients saw their tumours shrink.

What’s next?

Overall, I think these very promising results raise hopes that the phase II trial will also produce positive results at the American Society of Hematology (ASH) in December, which will be really great news for patients with Hodgkin Lymphoma.

If the phase II results also look positive, then we can probably expect Seattle Genetics and Millennium to file in the first half of 2011.

ResearchBlogging.org Younes, A., Bartlett, N., Leonard, J., Kennedy, D., Lynch, C., Sievers, E., & Forero-Torres, A. (2010). Brentuximab Vedotin (SGN-35) for Relapsed CD30-Positive Lymphomas New England Journal of Medicine, 363 (19), 1812-1821 DOI: 10.1056/NEJMoa1002965

3 Comments

Metastatic melanoma is quite a hot topic right now with a rich pipeline of products in development after a decade of little or no progress.  Of course, it is a bit like three London buses coming along at once after an hour long wait in the winter weather, but better late than never.

Many of you will remember the recent data from ipilimumab (BMS), an immunotherapy that showed increased survival, albeit with some severe adverse events, from the phase III trial in newly diagnosed metastatic melanoma presented at ASCO in the plenary session earlier this year, followed by a publication in the NEJM.  The FDA filing was subsequently submitted on the basis of the positive data.

Yesterday, BMS announced that the FDA have moved the PDUFA date back 3 months from Dec 25th to March 26th, 2011.  A precise reason for the delay wasn’t given , but the company did say:

“In response to an FDA request, Bristol-Myers Squibb submitted further analysis of data pertaining to the current application for pre-treated advanced melanoma and the agency considers this to be a major amendment to the drug’s BLA.”

I’m not going to speculate on the reasons for the extra review time or what the new data was, but it is an interesting and unexpected development.

Meanwhile, there’s also been a lot of buzz around targeted BRAF inhibition in melanoma lately, specifically around the initial stunning results seen with PLX4032 (Plexxikon & Roche).  So far, it seems that responses of around 6-12 months, with a median of around 8 months are possible with an kinase inhibitor that specifically targets the V600E mutation associated with BRAF, although there two problems:

  1. The responses are not durable as resistance (eg associated with MEK or AKT amplification) sets in.
  2. Inhibiting CRAF as well as BRAF appears to lead to an unwanted excess proliferation of squamous cells, which is reversible on withdrawal of treatment.

In the first case, a couple of recent papers have looked at mechanisms of resistance around BRAF inhibition that give us some clues of where to go next.

Gopal et al., (2010) decided to see what happened with AZD6244 or selumetinib (Array and AstraZeneca), a MEK and MAP/ERK inhibitor, and whether it would have any impact in mitigating BRAF resistance, given the potential close interaction within the RAS-RAF-MAPK pathway and downstream events that could be impacted through cross-talk and feedback loops:

“We analyzed a panel of Braf mutant human cutaneous melanoma cell lines for their sensitivity to growth and survival inhibition by AZD6244. We compared these effects with the baseline activation status of signaling pathways in the cells, and with AZD6244 treatment–induced changes in signaling networks.

These studies have identified the phosphoinositide 3-kinase (PI3K)-AKT pathway as a critical regulator of the efficacy of AZD6244 in Braf-mutant melanomas, including in cells without baseline activation of the pathway.”

In order to determine possible mechanisms of resistance in the cell lines, they compared the effects of AZD6244 treatment on their signaling pathways with effects in sensitive cell lines and found:

“Although all four of these Braf-mutant cell lines showed similar degree and duration of MAPK inhibition and several other proteins, the resistant cell lines increased their P-AKT levels following exposure to AZD6244, which was not observed in the sensitive cell lines.”

They went on to note:

“The functional significance of AKT activation is supported by the fact that inhibition of AKT activity, either by AKT knockdown or concurrent treatment with the mTORC1/2 inhibitor AZD8055, resulted in synergistic cell killing in the resistant cell lines.”

AstraZeneca and Merck have an ongoing partnership with their MEK (AZD6244) and AKT (MK-2206) kinase inhibitors, so combining them in a clinical trial to try and reduce resistance via feedback loops here would be an interesting approach worth trying.  Such a combination trial is currently recruiting in advanced solid tumours, not melanoma per se.  It is, however, a classic catch-all phase I study to see what kinds of cancers might respond and determine the MTD, but I would be very interested to see the data from patients with metastatic melanoma if they are enrolled.

Now, it has been shown in breast cancer cell lines showed that MEK inhibition resulted in cross-activation of the EGFR tyrosine growth factor receptor, but EGFR has not been shown to be relevant in melanoma, so Gopal et al., considered what other receptors might be responsible for mediating the effects.   In the discussion, an interesting snippet caught my eye:

“AZD6244 treatment induced a slight increase of IGF-I secretion by the cells, and knockdown of IGF-I also blocked P-AKT induction by AZD6244.  Supporting a specific role for the pathway in cell survival, recombinant IGF-I treatment blocked AZD6244-induced cell death, but not growth arrest, in the sensitive WM35.”

This might also suggest another useful combination approach to consider in clinical trials.

Previously, it has been shown that targeting BRAF can not only inhibit the important driver in melanoma, the V600E mutation, but it can also stimulate cellular signaling through the MEK-ERK pathway by activating the related family member C-RAF. This may explain the squamous cell proliferation seen in some patients with PLX4032. The more ideal BRAF inhibitor would therefore specifically target BRAF V600E, without activating CRAF at the same time.

Related to the subject of malignant melanoma, Kamata et al., (2010) just published a paper that looked at the relationship between BRAF and CRAF in the disease.  Previously it has been shown that D594A BRAF lacks kinase activity, but can induce the related gene product CRAF in addition to the mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK pathway.  What they found was really interesting.  In a nutshell:

“We show that the aneuploid phenotype is dependent on Craf. Treatment with the MEK inhibitor U0126 did not attenuate the emergence of aneuploidy but prevented the growth of aneuploid cells.  These results provide a previously unidentified link between Craf and chromosomal stability, with important implications for our understanding of the development of cancers with driver mutations that hyperactivate Craf.”

Aneuploidy is an abnormal number of chromosomes and can lead to genetic instability, a key cancer hallmark. It’s an important concept here because Kamata et al., have offered a different reason for the CRAF proliferation observed with some BRAF inhibitors:

“Impaired activity BRAF mutants are frequently coincident with oncogenic RAS mutations in human cancers (26) and in these, albeit rare, cancers, we may expect the hyper-activated CRAF induced by the combination of both oncogenes to enhance the aneuploidy response compared with mutation of either oncogene alone.  Such a situation is likely to be highly detrimental to the individual and, indeed, this mechanism may well account for the highly aggressive melanomas we observed following the combined expression of D594A Braf and G12D Kras in melanocytes.”

All in all, this is a very complex yet fascinating area of research and for those of you interested in this field, I would highly recommend reading the latest papers.

Photo Credit: Wikipedia

References:

ResearchBlogging.org Boni, A., Cogdill, A., Dang, P., Udayakumar, D., Njauw, C., Sloss, C., Ferrone, C., Flaherty, K., Lawrence, D., Fisher, D., Tsao, H., & Wargo, J. (2010). Selective BRAFV600E Inhibition Enhances T-Cell Recognition of Melanoma without Affecting Lymphocyte Function Cancer Research, 70 (13), 5213-5219 DOI: 10.1158/0008-5472.CAN-10-0118

 

Garnett MJ, Rana S, Paterson H, Barford D, & Marais R (2005). Wild-type and mutant B-RAF activate C-RAF through distinct mechanisms involving heterodimerization. Molecular cell, 20 (6), 963-9 PMID: 16364920

Gopal, Y., Deng, W., Woodman, S., Komurov, K., Ram, P., Smith, P., & Davies, M. (2010). Basal and Treatment-Induced Activation of AKT Mediates Resistance to Cell Death by AZD6244 (ARRY-142886) in Braf-Mutant Human Cutaneous Melanoma Cells Cancer Research, 70 (21), 8736-8747 DOI: 10.1158/0008-5472.CAN-10-0902

Kamata, T., Hussain, J., Giblett, S., Hayward, R., Marais, R., & Pritchard, C. (2010). BRAF Inactivation Drives Aneuploidy by Deregulating CRAF Cancer Research, 70 (21), 8475-8486 DOI: 10.1158/0008-5472.CAN-10-0603

4 Comments

Here’s a nice paper that I’ve been reading, written by Tim Harris and Frank McCormick on cancer biology.  Lately, we’ve all seen how advances in DNA sequencing and genome-wide association studies (GWAS) are driving the discovery of the germline and somatic mutations that are present in different cancers.  This article sets out to review:

“The most important molecular changes in different cancers from the perspective of what should be analyzed on a routine basis in the clinic.”

Essentially, this is an overview of where we are in both hematologic and solid tumours and looks at the molecular subsets that are emerging, hopefully as targets for therapeutic intervention.  I’m not going to repeat their excellent article, but if you are really interested in this field, I highly recommend reading it.  A link is provided below after the post to the actual paper.

What struck me most though, was not the nice summary of what we know about the biology of cancer, but their vision of the future in cancer medicine:

“The year is 2020. I wake up and feel the lump under my arm that has been bothering me for several weeks. I decide to make an appointment with the doctor to find the cause, especially since my personal genetic analysis has highlighted alleles that are associated with an increased risk of cancer.

The doctor’s receptionist views my electronic health record online before I am buzzed in to see the doctor.  Once I have explained the problem, a biopsy from the offending lymph-node is taken, the tissue is flash-frozen using the nitrogen quick-freeze system, and then delivered to the laboratory downstairs for a rapid molecular work-up.

DNA from the tissue is sequenced to identify any mutations in the 500 most common genes known to be involved in cancer. Tissue sections are analyzed using high-resolution fluorescent optical images.  A blood sample is also taken to check my background genomic DNA sequence, concentrating on alleles known to predispose to lymphoma.  Sequences for the genes encoding drug-metabolizing enzymes and drug-distribution proteins are also obtained.

A proteomic work-up is undertaken to look at protein profiles and post translational modifications.  I also undergo new-generation imaging so the gross pathology of my organs can be viewed in three-dimensions.  Two hours later, I review the results on my handheld computer device. The results have been predigested and presented as a simple digital read-out so that a diagnosis, prognosis and appropriate treatment can be derived.

Fortunately, the overall molecular and cellular pathology of the lymph-node is considered normal.  Apart from my pre-existing heart disease, all other organs appear to be healthy and I am prescribed an anti-inflammatory drug.

I leave the doctor’s office with a sigh of relief that all appears to be well.”

Whoa, that may sound like something from Bones in Star Trek initially, but I suspect it may well not be as far fetched as we imagine.  Why?   Because over the last two years the progress made in systems biology and cancer genome studies have encouraged me greatly.  What was a fledging area of cancer research is now becoming very much to the forefront of new breakthroughs and increased understanding of what is happening at the molecular level and new prognostic and predictive biomarkers are emerging.

Of course, there is still a long way to go in the war on cancer, but I see plenty of signs that much progress is being made.  Where we may well fall down though, is not in the science per se, but rather in our efforts of communication and coordination:

“The stakeholders, which include the payers, health care organizations, pharmaceutical and biotechnology industry, and molecular diagnostics companies, need to be aligned to achieve the most effective partnership.”

We clearly have much to do in this direction.  We have many of the electronic tools available already and yet speed of testing, diagnosis, access to electronic patient records (EHR) and in particular communication, is often snail mail slow.  How many of us receive test results instantly on our PDAs or email? How many have physicians or other service providers who even communicate with their patients by email or other technologies? This needs to change, and for the better.

There are some nice examples of real empowerment emerging from the cancer community, as this post from my friend Jody, a breast cancer survivor, shows in her recent blog post.  It’s a great start and I hope to hear of many more examples like this.

What do you think?   What can be done to improve the delivery of healthcare for people with cancer and how can we foster greater collaborations?

ResearchBlogging.org
Harris, T., & McCormick, F. (2010). The molecular pathology of cancer Nature Reviews Clinical Oncology, 7 (5), 251-265 DOI: 10.1038/nrclinonc.2010.41

8 Comments
error: Content is protected !!