Friday, November 27, 2009

Stapled peptides for targeting intracellular signaling pathways: research and commercial development

In the 12 November issue of Nature, there was a research article and a News and Views minireview about targeting an intracellular signaling pathway with a novel type of compound called a stapled peptide.

Signaling pathways are crucial for cellular physiology, and in the pathobiology of important diseases ranging from metabolic diseases to cancer. In many cases, signaling proteins that work by binding to other proteins in protein-protein interactions are key control points in signaling pathways. However, protein-protein interactions in all but a few cases cannot be readily addressed with small molecule drugs. These targets are therefore called “undruggable”. Some signaling pathways consist entirely of these “undruggable” targets, and can only be addressed indirectly (if at all) via targeting other pathways that interact with them.

Several small-molecule drugs that do address protein-protein interactions are natural products. The best known of these is the immunosuppressant FK506 (tacrolimus, Astellas’ Prograf). This is one reason for the new interest in natural products by some companies and researchers, as we discussed in a previous blog post.

However, the 12 November Nature article, authored by James E Bradner (Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA, and the Dana-Farber Cancer Institute, Boston, MA), Gregory Verdine (Department of Chemical Biology, Harvard University, Cambridge MA, and the Dana-Farber Cancer Institute), and their colleagues, takes a different approach. The researchers target specific intracellular protein-protein interactions by designing special types of peptides known as stapled peptides.

The signaling pathway that is the focus of this article is the Notch pathway. In normal physiology, this pathway regulates various aspects of cell-cell communication, cellular differentiation, cell proliferation, and cellular survival or death. Deregulated Notch pathway function is involved in diseases including cancers of the lung, ovary and pancreas, and in T-cell acute lymphoblastic leukemia (T-ALL), which is a cancer of immature T cells.

Notch is a cell-membrane receptor. Binding of one of its ligands (on the surface of an adjacent cell) to the extracellular domain of Notch triggers sequential cleavage of the Notch intracellular domain by a metalloproteinase known as TACE (tumor necrosis factor alpha converting enzyme) and by γ-secretase (an enzyme which is also involved in the amyloid pathway that is implicated in Alzheimer’s disease). The free intracellular domain of Notch, called ICN, migrates to the nucleus, and docks with the DNA-bound transcription factor CSL. The interaction between CSL and ICN creates a groove along the interface of the two proteins, which serves as a docking site for the mastermind-like protein MAML1. The resulting trimolecular complex initiates specific transcription of Notch-dependent target genes.

The binding domain of MAML1 that engages the elongated groove formed by the ICN-CSL complex is in the form of an α-helix. The researchers therefore designed a series of peptides derived from portions of the sequence of the MAML1 binding domain. These were stapled peptides in which hydrocarbon moieties are used to constrain, or “staple”, MAML1 binding-domain mimetic sequences into an α-helical conformation. One such stapled peptide, SAMH1, gave the highest affinity binding to ICN and CSL, and competitively inhibited binding of wild-type MAML1 to these proteins.

SAMH1 was cell-penetrant, and inhibited intracellular Notch pathway signaling in cultured T-ALL cell lines. Moreover, SAMH1 reduced the proliferation of a variety of T-ALL cell lines in vitro, but was inactive against T-cell tumor lines that were not dependent on the Notch pathway for their proliferation. In SAMH1-sensitive T-cell tumor lines, SAMH1 treatment activated caspases, which are involved in apoptosis. In a mouse model of T-ALL, intraperitoneally injected SAMH1 inhibited leukemic progression, and inhibited Notch pathway signaling in leukemic cells in vivo.

Stapled peptides are not conventional “drug-like” compounds. Their molecular weights are several times greater than the 500-dalton maximum prescribed by Lipinski’s rules (developed by the leading medicinal chemist Chris Lipinski), which are used to define “drug-like” properties of small molecule compounds. Moreover, peptides are usually subject to protease degradation in vivo, and thus have short serum half-lives. In most cases, peptides do not enter into cells efficiently, except for those peptides that have specific cell-membrane receptors.

However, stapled α-helical peptides, in addition to their improved binding activities to their specific targets, are protease-resistant, have improved serum half-lives, and are cell penetrant. Researchers attribute these properties to the constrained conformation of these molecules, and to the hydrocarbon staples themselves. For example, the hydrocarbon staples may confer lipophilic properties to these molecules, and thus render them membrane-penetrant.

In an earlier study, Dr. Verdine and researchers at the Dana-Farber Cancer Institute and Children’s Hospital in Boston designed a stapled α-helical peptide that initiated apoptosis by specifically binding to and activating a member of the Bcl-2 family, and that inhibited the grown of leukemic cells in a mouse model. The researchers have been continuing to develop and to determine the mechanisms of action of their Bcl-2 family-targeting stapled peptides.

The discovery-stage biotechnology company Aileron Therapeutics was founded in 2005 to develop and commercialize stapled peptides. The company’s scientific founders include Dr. Verdine, Loren Walensky (Dana-Farber Cancer Institute), and the late Stanley J. Korsmeyer (Dana-Farber Cancer Institute, a pioneer in the study of the Bcl-2 family and its role in apoptosis and in the biology of cancer). It has a pipeline of stapled peptides that it is developing for the treatment of solid and hematological tumors, the most advanced of which are in the preclinical stage. Aileron has managed to attract venture capital despite the current adverse conditions--in June 2009, the company closed a $40 million Series D financing.

Stapled peptides represent an exciting and innovative technology with the potential to address “undruggable” protein-protein interactions, and thus to treat diseases that represent major unmet medical needs. However, this technology is in an early stage, and the therapeutic value of stapled peptides has not yet been confirmed in the clinic.

Sunday, November 8, 2009

Anti-aging biology: new basic research, drug development, and organizational strategy

In the 2 October issue of Science (the "Ardipithecus ramidus issue”), there was a Perspective (authored by Matt Kaeberlein and Pankaj Kapahi) and a Report (authored by Colin Selman and his colleagues) on recent findings in anti-aging biology.

Since the late 1980s, researchers have found that caloric restriction (CR) (reduction in caloric intake while maintaining essential nutrients) slows aging in a variety of organisms—yeasts, nematodes, fruit flies, mice, and most recently rhesus macaques. In the recently published 20-year study in rhesus macaques, CR not only increased lifespan, but also delayed the onset of a suite of aging-related disease conditions—diabetes, cancer, cardiovascular disease, and brain atrophy. This parallels the studies with other organisms.

Researchers who have been studying the CR model have been attempting to elucidate the mechanisms by which CR works to slow the aging process and to retard aging-related disease. They hope to find targets for drugs to mimic the effects of CR in humans, since long-term CR is not practical for most people. Over the years, researchers have discovered several pathways by which CR appears to exert its effects. The Report describes new research results on one such pathway, the mammalian target of rapamycin (mTOR) pathway. The Perspective reviews this research in the context of related recent studies.

In a report published in Nature earlier this year (16 July 2009), researchers found that rapamycin administered in food increased the median and maximal lifespan of genetically heterogeneous laboratory mice, whether it was fed to middle-aged (600 days old) or young adult (270 days old) mice. Rapamycin feeding beginning at 600 days of age led to an increase in lifespan of 14% for females and 9% for males, on the basis of age at 90% mortality.

Rapamycin targets mTOR (mammalian target of rapamycin), a kinase that regulates signaling pathways that affect many cellular processes. mTOR forms two protein complexes that are active in intracellular signaling—mTORC1 and mTORC2. It is mTORC1 that is most sensitive to rapamycin. mTORC1 works to coordinate cellular growth and survival responses induced by changes in the availability of nutrients, and also responses to cellular stresses (e.g., hypoxia, DNA damage and osmotic stress). Genetic inhibition of TORC1 in yeast and invertebrates has been found to extend their lifespan. In particular, in the nematode Caenorhabditis elegans, TORC1 interacts with the insulin pathway (via raptor, a component of TORC1) to control lifespan. The role of the insulin pathway in the enhancement of lifespan by CR in C. elegans has been known for many years. The role of mTORC1 at the junction of nutrient and stress sensing pathways, together with these results in invertebrates and now mice, has led researchers to hypothesize that the mTORC1 pathway may be involved in CR-mediated enhancement of lifespan, and that drugs that modulate this pathway may substitute for CR in lifespan extension.

In other studies, inhibition of the mTOR pathway in mice was found to retard development of such aging-related conditions as cancer, metabolic disease, and cardiovascular disease. This effect has also been seen in studies of CR in mice and in nonhuman primates, as stated above.

Rapamycin is an immunosuppressant that is marketed as Wyeth’s (now Pfizer’s, since the October 2009 merger) Rapimmune, to prevent organ transplant rejection. More recently, a derivative of rapamycin, temsirolimus (Wyeth/Pfizer’s Toricel) has been approved for treatment of renal cell carcinoma. The authors of the Nature paper therefore hypothesized that rapamycin may have extended lifespan in the mice either by working via CR-related pathways that control lifespan, by postponing death from cancer, or both.

The finding that oral rapamycin can retard aging in mice, even when fed to 600-day-old mice (the equivalent of 60 years old in humans) raises hope for the development of anti-aging drugs for human use. However, rapamycin itself cannot be used for this purpose because of its immunosuppressant effects. (In the mouse rapamycin feeding studies, the mice were kept under specific pathogen-free conditions.) If researchers were to attempt to modulate the mTORC1 pathway to extend lifespan, they would therefore need to discover other drugs that modulate that pathway without rapamycin’s side effects. Learning more about specific pathway components that may be targeted to increase lifespan may help researchers discover such drugs.

In the new Selman et al. report, researchers endeavored to learn more about how the mTORC1 pathway might extend lifespan in mice. They constructed knockout mice that lacked S6 protein kinase 1 (S6K1). S6K1 is a downstream target of mTORC1, which upregulates mRNA translation and protein synthesis in response to mTORC1 signaling. The researchers found that deletion of the gene for S6K1 resulted in a 19% increase in median lifespan in female mice (as compared to wild-type females), and also increased maximum lifespan. S6K1 deletion had no effect on the lifespan of male mice. This was in contrast to the study with rapamycin feeding, which showed lifespan extension in both sexes, even though the effect in female mice was greater. However, the results of the two studies are not strictly comparable, since mice of different genetic background were used in the two studies.

Female S6K1 knockout mice also showed improvement in several biomarkers of aging (e.g., motor and neurological function, level of physical activity, insulin sensitivity, glucose tolerance, fat mass, immunological parameters). Hepatic gene expression in 600-day-old female S6K1 knockout mice resembled that of wild type mice subjected to CR. Female S6K1 knockout mice showed increased hepatic, muscle, and adipose tissue expression (as compared to wild-type mice) of genes associated with other pathways associated with longevity, including genes for sirtuin-1 (SIRT1) and adenosine monophosphate-activated protein kinase (AMPK).

Selman et al. went on to obtain evidence that the effect of S6K1 knockout on lifespan in female mice is due to activation of AMPK. The gene expression profile of muscle tissue of long-lived female S6K1 knockout mice resembled the profile of wild-type mice treated with the AMPK activator aminoimidazole carboxamide ribonucleotide (AICAR). Hepatocytes from S6K1 knockout mice also showed enhanced AICAR activation of AMPK as compared to hepatocytes from wild type mice. A parallel study in C. elegans showed that deletion of the aak-2 gene, which encodes a subunit of AMPK, suppresses lifespan extension in mutants that lack rsks-1, the nematode homolog of S6K1. These results suggest that S6K1 knockout may exert its pro-longevity effects via activation of AMPK.

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. AMPK has been implicated in metabolic response to CR in eukaryotic organisms from yeasts to humans, and it mediates the effects on lifespan of at least one type of CR regimen in C. elegans. Thus the hypothesis that lifespan extension via the mTORC1-S6K1 pathway works via AMPK activation is an attractive one.

However, it is not known how deletion of S6K1 (or its inhibition via mTORC1 in rapamycin-treated mice) might activate AMPK. Moreover, as pointed out by Kaeberlein and Kapahi, there are other downstream targets of S6K1 that might play a role in anti-aging effects of SK61 deletion or inhibition. Among these is hypoxia-inducible factor-1α (HIF-1α). Moreover, there are other biomolecules and pathways that have been implicated in the effects of CR on retarding aging. These especially include the sirtuins, an evolutionarily conserved family of nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylases.

As shown by the Perspective and Report in the 2 October issue of Science, anti-aging research is an exciting area of basic biological research, and researchers still have much to learn about pathways that mediate the effects of CR on longevity. However, this field is already being applied to drug discovery and development. A basic issue in applying anti-aging research to the development of drugs is that one clearly cannot use increased lifespan as an endpoint in clinical trials. Companies must test putative anti-aging drugs against one or more diseases of aging. The hope is that any “anti-aging” drugs approved for treatment of one disease of aging will have pleiotropic effects on multiple diseases of aging, and will ultimately be found to increase lifespan or “healthspan” (the length of a person’s life in which he/she is generally healthy and not debilitated by chronic diseases).

The two principal types of “anti-aging” drugs currently in company pipelines are sirtuin modulators and AMPK activators. Sirtris Pharmaceuticals (Cambridge, MA, a wholly-owned subsidiary of GlaxoSmithKline [GSK]) is developing the SIRT1 activators SRT501 (a proprietary formulation of the natural product resveratrol) and SRT2104 (a novel synthetic small-molecule SIRT1 activator that is structurally unrelated to resveratrol and is up to 1000-fold more potent). SRT501 is in Phase II clinical trials in type 2 diabetes. SRT2104 has been tested in Phase I trials in healthy volunteers, and was found to be safe and well tolerated. Elixir Pharmaceuticals (Cambridge, MA) is developing a preclinical-stage SIRT1 inhibitor for treatment of Huntington’s disease and certain cancers, and a preclinical-stage SIRT1 activator for treatment of type 2 diabetes and obesity. Elixir also has a research-stage SIRT2 inhibitor under development for treatment of type 2 diabetes and obesity.

Companies developing AMPK activators include a collaboration between Metabasis Therapeutics (La Jolla, CA; about to be acquired by Ligand Pharmaceuticals, San Diego, CA) and Merck--preclinical oral AMPK activators, for treatment of type 2 diabetes and hyperlipidemia), Mercury Therapeutics (Woburn, MA)--research and preclinical-stage oral AMPK activators for treatment of type 2 diabetes, and Betagenon (Umea, Sweden)--the preclinical-stage oral AMPK activator BG8702, for treatment of type 2 diabetes.

The relationship between sirtuin-modulator developer Sirtris and GSK represents a prime example of the attempt of large pharmaceutical companies to become more “biotech-like” in order to improve their R&D performance. We discussed this strategy in our recent report, Approaches to Reducing Phase II Attrition. GSK acquired Sirtris for $720 million in June 2008. In December 2008, GSK announced that it had appointed Christoph Westphal, the CEO and co-founder of Sirtris, as the Senior Vice President of GSK’s Centre of Excellence in External Drug Discovery (CEEDD). The CEEDD works to develop external alliances with biotech companies, with the goal of acquiring promising new drug candidates for GSK’s pipeline. Michelle Dipp, who was the vice president of business development at Sirtris at the time of GSK’s appointment of Dr. Wesphal, is now Vice President and the head of the US CEEDD at GSK. Dr. Westphal, who is also a former venture capitalist, remains as CEO of Sirtris, and is based at Sirtris’ Cambridge location.

Thus anti-aging research, despite the fact that it is mainly in the basic research stage, is not only beginning to produce drug candidates, but has also been having an impact on the organizational strategy of one of the major pharmaceutical companies, GSK.