19 May 2011

Can HDL-raising drugs be a big field after all?

By |2018-09-14T21:57:22+00:00May 19, 2011|Cardiovascular Disease, Drug Development, Strategy and Consulting, Translational Medicine|

 

Atherosclerosis. From Nephron. http://bit.ly/jL6Zos

 

In the April 29, 2011 issue of Cell, there is a Leading Edge review entitled “Macrophages in the Pathogenesis of Atherosclerosis”, by Kathryn J. Moore (New York University Medical Center, New York, NY ) and Ira Tabas (Columbia University, New York, NY). This 15-page  review (including 4 pages of references) covers a big subject–the central role of the macrophage in the pathogenesis of atherosclerosis, and of the resulting acute thrombotic vascular disease, including myocardial infarction, stroke, and sudden cardiac death. The review will be helpful to those who wish to update their knowledge of the mechanistic basis of atherothrombotic disease, or to those who want an introduction to the subject.

Included in the review is a discussion of the role of high-density lipoprotein (HDL), or “good cholesterol” in promoting regression of atherosclerotic plaques. HDL, as well as the protein component of HDL, apolipoprotein A1, are key players in the process of cholesterol efflux, or removal of cholesterol from macrophages in atherosclerotic plaques. HDL may also have other beneficial roles, including prevention of subendothelial apolipoprotein B-lipoprotein (apoB-LP) retention (which starts the atherosclerotic process in the first place), decreasing activation of endothelial cells, and reducing LDL oxidation. (ApoB is the protein component of low-density lipoprotein [LDL], or “bad cholesterol”.) In human populations, low HDL is generally recognized as a major cardiovascular risk factor, and high HDL is recognized as being protective.

In its discussion of therapeutic strategies based on our current picture of the mechanistic basis of atherosclerosis, the authors of the review state that the most effective way to treat the condition would be to decrease subendothelial apoB-LP retention by lowering apoB-LPs in the blood via lifestyle changes and drugs. In order to completely prevent atherosclerosis, serum apoB-LPs (i.e., mainly LDL and VLDL [very low-density lipoprotein]) would need to be lowered below the threshold level required for subendothelial apoB-LP retention in the arteries. However, in Western societies (and in other societies that have been rapidly adopting Western lifestyles), initiation of atherosclerotic lesions occurs in the early teens; thus this preventive approach is not currently feasible.

The leading drugs for lowering serum LDL are the statins, such as atorvastatin (Pfizer’s LIpitor, which is the largest-selling statin; Lipitor will go off-patent in November 2011), pravastatin (Bristol-Myers Squibb’s Pravachol, generics), simvastatin (Merck’s Zocor, generics), and rosuvastatin (AstraZeneca’s Crestor). Statins are generally accepted as being effective in decreasing mortality in patients with cardiovascular disease (CVD). These drugs are also widely prescribed for patients with a high risk of developing CVD; i.e., patients with high LDL, type 2 diabetes, and/or other risk factors. However, some researchers question the value of statins in primary prevention in patients without preexisting CVD but at high risk of developing the disease. For example, a 2010 meta-analysis published in the Archives of Internal Medicine did not find evidence that statin therapy was beneficial in primary prevention of all-cause mortality in patients at high risk of developing CVD. Moreover, although statins are highly effective in decreasing cardiovascular events (up to 60%) and cardiovascular deaths in patients with pre-existing CVD, a large percentage of patients with or at high risk of developing CVD, despite statin treatment, still experience cardiovascular events and cardiovascular death. Therefore, researchers and companies would like to develop other, complementary drugs that work via different mechanisms from the statins.

HDL raising has long been a key target for pharmaceutical and biotechnology companies in their quest to develop CVD drugs that would be complementary to the statins. In the early-to-mid 2000’s, companies had several candidate drugs, of different types, in development. In an article published by Pharmaceutical Executive in 2006, I was quoted as saying that raising HDL was a big field. However, most of the drugs being developed at that time fell by the wayside, mainly due to failure in the clinic.

A particular focus of pharmaceutical companies has been the development of cholesteryl ester transfer protein (CETP) inhibitors. CETP catalyzes the transfer of cholesteryl esters and triglycerides between LDL/VLDL and HDL, and vice versa. In vivo (in animals and in humans), CETP inhibitor drugs raise HDL and lower LDL.

The leading CETP inhibitor in the early to mid-2000s was Pfizer’s torcetrapib. Pfizer had placed high hopes on torcetrapib, as a potential blockbuster to replace anticipated lost revenues from Lipitor when it went off-patent in 2011. However, in late 2006 Pfizer pulled the drug from Phase 3 trials, after finding that combination therapy with torcetrapib  and atorvastatin gave a 50 percent greater mortality rate that atrovastatin alone. This was not only a huge disappointment for Pfizer and its shareholders, but also cast a pall of gloom over the entire HDL-raising drug field, and especially over CETP inhibitors. Researchers speculated that inhibition of CETP might result in producing a form of HDL that is not cardioprotective, and might even be harmful. There were even calls for pushing the HDL field back to the basic research level, with the need to find just how (and what form of) HDL exerted its cardioprotective effects, in people with elevated HDL due to genetics, lifestyle, or treatment with high-dose niacin (the only drug approved to raise HDL).

However, later studies of torceptrapib found that the toxicity of the compound was not due to an untoward effect of CETP inhibition or HDL raising, but was due to off-target effects of the drug. In animals and in humans, torceptrapib raised serum levels of aldosterone, via release of aldosterone from the adrenals. Aldosterone was responsible for the increase of blood pressure seen in animals and in humans treated with torceptrapib, and aldosterone has proatherogenic effects that go beyond its effects on blood pressure. The hypertensive and aldosterone-raising effects of torceptrapib were independent of its CETP inhibitor activity, and other CETP inhibitors (discussed below) do not raise aldosterone levels or blood pressure.

A March 2011 News and Analysis article in Nature Reviews Drug Discovery reviewed the history of the CETP inhibitor field after the demise of torcetrapib. Although the torcetrapib debacle caused several other companies to exit the CETP inhibitor field, Roche and Merck persisted. Roche has been developing the CETP inhibitor  dalcetrapib, and Merck’s CETP inhibitor is known as anacetrapib.

As mentioned in the Nature Reviews Drug Discovery mini-review, Dr. Alan Tall (Columbia University), working in collaboration with Merck researchers, showed in 2010 that niacin treatment in humans resulted in a 30% increase in HDL, while anacetrapib treatment resulted in a 100% increase in HDL. Niacin treatment in humans resulted in a moderate increase in the ability of HDL to promote net cholesterol efflux (measured in in vitro assays) while anacetrapib treatment caused a more dramatic increase. This was due not only to a higher level of HDL in anacetrapib-treated subjects, but also to enhanced ability of anacetrapib-induced HDL particles to promote cholesterol efflux, especially at high HDL concentrations. HDL from both niacin-treated and anacetrapib-treated subjects also exhibited anti-inflammatory activity. This study should help lay to rest the idea that pharmacological inhibition of CETP might result in abnormal pro-atherogenic HDL, as theorized by some researchers after the clinical failure of torceptrapib.

Currently, Roche’s dalcetrapib is in a 15,600-patient Phase 3 clinical trial known as dal-OUTCOMES; this trial was initiated in 2008, and efficacy results are expected in 2012-2013. As of the time of the Nature Reviews Drug Discovery article, Merck planned to initiate its 30,000-patient REVEAL trial of anacetrapib in April 2011. Efficacy results of REVEAL are anticipated in 2014-2016.

In December 2010, the results of Merck’s moderate-sized (1623 patients with or at high risk for CVD, who were already being treated with a statin) Phase 3 DEFINE trial of anacetrapib were published in the New England Journal of Medicine. The DEFINE trial was designed as a safety study. In this 76-week study, anacetrapib showed no significant differences from placebo in terms of safety, as measured by a pre-specified cardiovascular endpoint (defined as cardiovascular death, myocardial infarction, unstable angina or stroke). These cardiovascular events occurred in 16 anacetrapib-treated patients (2.0 percent) compared with 21 placebo-treated patients (2.6 percent). There were also no significant differences in blood pressure, serum electrolytes, or aldosterone levels between anacetrapib-treated and placebo-treated patients.

Anacetrapib treatment also decreased LDL by 40 percent (from 81 to 45 mg/dl vs. 82 to 77 mg/dl for placebo) and increased HDL by 138 percent (from 40 to 101 mg/dl vs. 40 to 46 mg/dl for placebo). Anacetrapib also had other favorable effects on lipid levels (e.g., 36.4% reduction in lipoprotein(a), and 6.8% reduction in triglycerides, beyond the changes seen with placebo treatment).

Although the DEFINE study was too small to provide definitive results regarding the safety of anacetrapib, it gave a 94% predictive probability that treatment with anacetrapib is not associated with the rate of cardiovascular events seen with torcetrapib. The trial also indicated that anacetrapib treatment does not result in the effects (especially raising of serum aldosterone levels) thought to be responsible for torcetrapib’s toxicity. Moreover, anacetrapib treatment resulted in a dramatic increase in HDL levels (beyond that seen with torcetrapib) in the DEFINE study, and the 2010 study by Dr. Tall and his colleagues indicates that anacetrapib-induced HDL is highly effective in promoting cholesterol efflux.

The results with anacetrapib have reignited enthusiasm for CETP inhibitors in the medical community. Even the often-critical Dr. Steven Nissen (Cleveland Clinic) expressed enthusiasm for anacetrapib. However, despite these promising results, the efficacy of CETP inhibitors, in terms of significantly reducing the rate of cardiovascular events, has not yet been demonstrated. Only large, adequately-powered Phase 3 clinical trials, such as dal-OUTCOMES for Roche’s dalcetrapib and REVEAL for Merck’s anacetrapib, can definitively establish both the efficacy and the safety of these drugs.

The development of CETP inhibitors represents a situation in which the leading drug in the class failed because of off-target effects. However, these off-target effects were not class effects, and targeting CETP in order to raise HDL now seems like a good idea after all. Pfizer ignored warning signs (especially the modest elevation in blood pressure induced by torcetrapib, which did not appear to be very significant) in pursuit of its commercial goals, while Roche and especially Merck pursued a more moderate and science-based approach to development of CETP inhibitors. Other companies stopped development of their CETP inhibitors, thus losing their opportunities in this field. Meanwhile, various companies and academic group have been developing other approaches to HDL raising, such as apolipoprotein A1 mimetics, which are in early stages of development.

Despite its early setbacks, HDL-raising drugs may turn out to be a big field after all.

_____________________________

As the producers of this blog, and as consultants to the biotechnology and pharmaceutical industry, Haberman Associates would like to hear from you. If you are in a biotech or pharmaceutical company, and would like a 15-20-minute, no-obligation telephone discussion of issues raised by this or other blog articles, or of other issues that are important to  your company, please click here. We also welcome your comments on this or any other article on this blog.

13 May 2011

Workshop on improved animal models for pharma R&D at the World Drug Targets Summit, July 2011

By |2011-05-13T00:00:00+00:00May 13, 2011|Animal Models, Cancer, Drug Development, Drug Discovery, Haberman Associates, Neurodegenerative Diseases, Strategy and Consulting|

 

I will lead a workshop entitled “Developing Improved Animal Models in Oncology and CNS Diseases to Increase Drug Discovery and Development Capabilities” at the World Drug Targets Summit in Cambridge MA in July 2011.

Workshops will be held on July 19, and the main conference on July 20-21. I am planning to attend the entire conference.

Our workshop will be a discussion of 2-3 case studies involving development of novel animal models in oncology and CNS diseases, aimed at more closely modeling human disease than current models. Drug discovery and development in these therapeutic areas has been severely hampered by animal models that are  poorly predictive of efficacy. This is a major cause of clinical attrition in these areas.

We shall discuss the implications of these case studies for developing novel therapeutic strategies, target identification and validation, drug discovery, preclinical studies, and reducing clinical attrition. We shall also discuss hurdles to industry adoption of novel animal models developed in academic laboratories.

The main conference will focus on ways of building successful target strategies to minimize drug attrition in the clinic, and specifically how to identify and validate targets that can lead to commercially differentiated products. Speakers will include target discovery and validation leaders from such companies as Pfizer, Merck, NeurAxon, Gilead Sciences, Boehringer Ingelheim, Merrimack Pharmaceuticals, Bayer Schering Pharma AG, FORMA Therapeutics, Roche, Novartis, Tempero Pharmaceuticals, UCB Pharma, Infinity Pharmaceuticals, and from such academic institutions as Harvard Medical School.

The conference agenda and brochure, as well as online registration, are available on the conference website.

3 May 2011

The great metformin mystery–genomics, diabetes, and cancer

By |2018-09-12T21:46:50+00:00May 3, 2011|Anti-Aging, Cancer, Drug Development, Drug Discovery, Metabolic diseases, Strategy and Consulting|

 

Galega officinalis (Goat’s Rue) From JoJan http://bit.ly/l5Ybco

Metformin (Bristol-Myers Squibb’s Glucophage, generics), an oral biguanide antidiabetic drug, is the most widely prescribed agent for treatment of type 2 diabetes. The drug mainly works by lowering glucose production by the liver, and thus lowering fasting blood glucose.

Although metformin–approved in the United States in 1994, and in Europe prior to that–has been used for many years, its mechanism of action is not well understood. In 2005, signal-transduction pioneer Lewis Cantley (Beth Israel Deaconess Cancer Center/Harvard Medical School, Boston MA), and his colleagues–including Reuben J. Shaw (now at the Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA)–published a report showing that metformin targets the adenosine monophosphate (AMP)-activated kinase (AMPK) pathway in the liver. We discussed this report and its implications in our 2007 Cambridge Healthtech Institute Insight Pharma Report, Diabetes and Its Complications.

AMPK is found in all eukaryotic organisms, and serves as a sensor of intracellular energy status. In mammals, it also is involved in maintaining whole-body energy balance, and helps regulate food intake and body weight. We  have discussed the potential role of AMPK in regulation of lifespan, and as a target in anti-aging medicine and in metabolic disease, in earlier articles on this blog. (See here and here.)

AMPK is activated by increases in the ratio of AMP to ATP, caused by energy stress. Under conditions of energy stress, AMP levels go up, and AMP binds to a specific site on the AMPK γ subunit. This induces a conformational change that exposes the activation loop of the α subunit. This allows an upstream serine/threonine kinase to phosphorylate this activation loop. In several mammalian cell types, including liver and skeletal muscle, that kinase is LKB1. Drs. Cantley and Shaw in 2005 showed that metformin targets the LKB1-AMPK pathway in the liver, and that metformin requires LKB1 to lower glucose production by the liver. However, neither LKB1 nor AMPK is the direct target of metformin, and as of 2005, that direct target was unknown.

A new genetic study that suggests that ATM kinase may affect the ability of patients to respond to metformin

Now–as of February 2011–comes a Nature Genetics paper that indicates that the serine/threonine kinase ATM (ataxia telangiectasia mutated) acts upstream of AMPK to mediate the therapeutic effects of metformin. ATM is a DNA repair protein that is recruited and activated by double-strand breaks in DNA. It initiates activation of the DNA damage checkpoint, leading to cell cycle arrest, followed by DNA repair or apoptosis. Thus the role of ATM in the AMPK pathway and in the therapeutic effects of metformin is surprising indeed.

In the study reported in the Nature Genetics paper, researchers of The GoDARTS and UKPDS Diabetes Pharmacogenetics Study Group and The Wellcome Trust Case Control Consortium 2 performed a genome-wide association study (GWAS) for glycemic response to metformin in type 2 diabetes patients in the U.K. In a population of nearly 4,000 patients, they identified a single-nucleotide polymorphism (SNP) designated rs11212617, which was associated with treatment success. This SNP occurs in a genetic locus that also contains the gene that encodes ATM. In a rat hepatoma cell line, inhibition of ATM by the specific inhibitor KU-55933 (KuDOS Pharmaceuticals, Cambridge, U.K., which was acquired by AstraZeneca in 2005) attenuated metformin-mediated phosphorylation and activation of AMPK.

The analysis by Morris Birnbaum and Reuben Shaw in the 17 February 2011 issue of Nature

The 17 February 2011 issue of Nature contained a Forum entitled “Genomics: Drugs, diabetes and cancer.” This consisted of two analyses of the implications of the Nature Genetics paper for metformin’s mechanism of action, and for understanding diabetes and the connections of the metformin-activated ATM/AMPK pathway with cancer. The first analysis was by Morris J. Birnbaum, M.D., Ph.D. (University of Pennsylvania Medical School, Philadelphia, PA), who does research on the role of AMPK and insulin in energy metabolism and in diabetes. The second analysis is by Dr. Reuben Shaw, mentioned earlier. Dr. Shaw’s research centers around LKB1 [also known as serine/threonine kinase 11 (STK11)]. LKB1, a serine/threonine kinase, is not only a regulator of hepatic glucose production via AMPK, but is also a tumor suppressor. Germline mutations in LKB1 are associated with the familial cancer Peutz-Jegher syndrome, and somatic mutations in LKB1 are present in various other cancers. In particular, the Lkb1 gene is one of the most frequently muted genes in human lung adenocarcinomas.

Dr. Birnbaum’s analysis

Dr. Birnbaum notes that the finding of a role for ATM in metformin responsiveness may be an important clue to the mechanism of action of this drug. However, it may also be a false lead, with ATM having only an indirect effect on metformin’s action. He cites recent evidence that metformin acts independently from LKB1 and AMPK and of transcriptional regulation in general. In these studies, genetic ablation of LKB1 and AMPK was used to show that these mediators are dispensable for metformin’s glucose-lowering activity. Instead, metformin appears to work by inhibiting mitochondrial production of ATP in the liver, thus reducing the level of liver glucose production via gluconeogenesis (which uses ATP). This is in apparent contradiction to the 2005 results of Dr Shaw and his colleagues. Nevertheless, metformin’s inhibition of mitochondrial ATP production increases the ratio of AMP to ATP, and thus activates AMPK. There are also other pathways by which inhibition of mitochondrial ATP production may inhibit gluconeogenesis. Thus the mechanisms by which metformin causes a decrease in glucose production by the liver appear to be very complex, and are not well understood.

Dr. Birnbaum therefore speculates that ATM may affect blood glucose levels via pathways that are parallel to, but not the same as, those modulated by metformin. However, the effects of these other pathways may be synergistic with those modulated by metformin when patients are treated with the drug. Dr. Birnbaum notes that 40 years ago, it was found that patients with ataxia telangiectasia often display a type 2-diabetes-like condition, including insulin resistance. Ataxia telangiectasia is a familial disease caused by germline mutations in ATM. This suggests that  ATM may act to counteract hyperglycemia and insulin resistance.

Dr. Birnbaum concluded that biochemical and cell biology studies should be conducted to determine the nature of the interaction of ATM and the antidiabetic effects of metformin. Key to these endeavors is to determine whether there are any biomolecules other than AMPK that both are influenced by ATM and control metabolism.

Dr. Shaw’s analysis

Dr. Shaw first discusses several animal studies that help elucidate the role in glucose regulation of the biomolecules involved in the putative ATM-LKB1-AMPK pathway. He notes notes that deletion of the Lkb1 gene in mouse liver results in loss of AMPK activity in that organ, and to the development of hyperglycemia and hepatic steatosis–two conditions that are seen in type 2 diabetes. Dr. Shaw also cites the 40-year-old finding about the connection between  ataxia telangiectasia and insulin resistance and diabetes. But as he also mentions the more recent (2006) finding that mice with defective ATM activity show increased insulin resistance and abnormal glucose regulation.

Dr. Shaw then speculates as to how ATM might work to modulate patients’ antidiabetic responses to metformin. He notes that ATM is known to phosphorylate LKB1, which is the key activator of AMPK in the liver. Alternatively, ATM might also regulate AMPK independently of LKB1, and might affect responsiveness of patients to metformin by regulating other relevant targets, independently of AMPK. In this context, ATM is known to phosphorylate other, LKB1 and AMPK-independent components of the insulin signaling pathway.

In the light of these considerations, Dr. Shaw says that it is important to determine whether the rs11212617 genetic variant results in modulation of ATM activity toward AMPK activation or toward other targets relevant to glucose regulation, or indeed whether this SNP affects ATM activity at all.

Dr. Shaw then focuses on the potential relevance of metformin to cancer therapy. Researchers have found, in retrospective studies, that diabetes patients who take metformin have a lower risk of developing cancer than those treated with other antidiabetic medications. Animal studies confirm the anticancer effects of metformin, but–as discussed in a 2010 review by Dr. Michael Pollak (McGill University, Montreal, Quebec, Canada)–they indicate that the anticancer effects of this drug are mechanistically complex. Dr. Shaw asks whether metformin is a general activator of ATM (and/or its targets) in the DNA damage-response pathway, or whether its specific effects on LKB1 and/or AMPK might be responsible for the apparent beneficial effects of metformin on cancer risk.

Dr. Shaw concludes with the statement that future studies of the relationship between metformin action, ATM, LKB1, and AMPK should shed light on the relationship between metformin’s antidiabetic effects and its apparent anticancer effects.

Our conclusions

The finding, based on a genome-wide association study, which suggests that ATM, a kinase best known for its involvement in DNA repair pathways, may also be involved in diabetics’ response to metformin is surprising and intriguing. It may eventually be important in unraveling metformin’s mechanism of action in inhibition of liver gluconeogenesis, and in other antidiabetic activities. This finding indicates a connection between pathways by which metformin exerts its antidiabetic activities, and pathways that are involved in cancer.

Nevertheless, the elucidation of metformin’s mechanism(s) of action in diabetes remains a work in progress. This situation is an example of how science works in the real world (as opposed to textbooks or much of science journalism)–generating more questions than answers.

A drug like metformin, with its complex and still poorly understood mechanism of action, could not have been discovered by modern, post-genomics drug discovery strategies. Metformin was discovered via research on natural products derived from the plant Galega officinalis (known as the French lilac, goat’s rue, and by various other names), which had been known by herbalists for centuries. It is fortunate that researchers were able to study the effects of extracts of this plant, and ultimately to develop metformin, well in advance of the modern era of drug discovery. Diabetics and their physicians now have access to metformin as an inexpensive generic drug.

The continued study of the antidiabetic mechanism(s) of action of metformin may yield additional insights into control of gluconeogenesis and other metabolic pathways. Some of the findings of these studies might be relevant to drug discovery and development, for example the development and use of AMPK activators in metabolic disease and in anti-aging medicine.

Continued study of the mechanism(s) of action of metformin may also be relevant to developing new therapies for cancer. As suggested by Dr. Pollak, although metformin is off-patent and is thus not an attractive agent for development as an oncology drug by pharmaceutical or biotechnology companies, other biguanides or related compounds might be better anticancer compounds, and would be patentable. In addition to identifying such compounds, it will be important to determine and define which groups of cancer patients could best benefit from them (perhaps via biomarkers). It will then be important to conduct personalized medicine hypothesis-testing clinical trials (as discussed in an earlier blog post) designed to obtain proof-of-concept that such compounds can indeed benefit specific groups of patients.

___________________________________

As the producers of this blog, and as consultants to the biotechnology and pharmaceutical industry, Haberman Associates would like to hear from you. If you are in a biotech or pharmaceutical company, and would like a 15-20-minute, no-obligation telephone discussion of issues raised by this or other blog articles, or of other issues that are important to  your company, please click here. We also welcome your comments on this or any other article on this blog.

Go to Top