30 June 2012

Lorcaserin (Arena/Eisai’s Belviq) approved by the FDA–the first new weight-loss drug in 13 years

By |2018-09-19T20:00:09+00:00June 30, 2012|Drug Development, Metabolic diseases|

 

Lorcaserin. Source: PubChem

On June 27, 2012, Arena Pharmaceuticals and its commercialization pattern Eisai, Inc. (Woodcliff Lake, NJ) announced that the U.S. FDA had approved its antiobesity drug lorcaserin–the first drug for long-term weight loss to be approved in the U.S. in 13 years. Lorcaserin will be marketed under the trade name Belviq.

The FDA approved lorcaserin as an adjunct to diet and exercise for chronic weight management in adult patients who are obese [initial body mass index (BMI) of 30 kg/m2 or greater], as well as for overweight patients with a BMI of 27 kg/m2 or greater who also have at least one weight-related comorbidity, such as hypertension, dyslipidemia, or type 2 diabetes. The approved indication includes a statement that the safety and efficacy of coadministration of lorcaserin with other products intended for weight loss and the effect of lorcaserin on cardiovascular morbidity and mortality have not been established.

According to the Arena/Eisai announcement, three double-blind, randomized, placebo-controlled trials showed that lorcaserin plus diet and exercise was more effective than diet and exercise alone in helping patients lose 5% or more of their body weight after one year and managing the weight loss for up to two years.

The most common adverse effects seen in nondiabetics treated with lorcaserin were headache, dizziness, fatigue, nausea, dry mouth, and constipation. In patients with type 2 diabetes, the most common adverse effects were hypoglycemia, headache, back pain, cough, and fatigue.

The FDA has recommended that lorcaserin be classified as a scheduled drug. The U.S. Drug Enforcement Administration (DEA) will review this recommendation and determine the final scheduling designation. Once this has been done, Eisai will announce when and under what terms lorcaserin will be available to U.S. physicians and patients.

The approval of lorcaserin includes a commitment by Arena and Eisai to conduct post-marketing studies to assess the safety and efficacy of lorcaserin for weight management in obese pediatric patients, as well as to evaluate the effect of long-term treatment with lorcaserin on the incidence of major adverse cardiovascular events in overweight and obese subjects with cardiovascular disease or multiple cardiovascular risk factors. The cardiovascular outcomes trial will include echocardiographic assessments.

The implications of the approval of lorcaserin for the obesity drug market

Arena is to be congratulated for its persistence in getting lorcaserin approved. As of February 1, 2011, all three of the obesity drug candidates that came up for FDA review in 2010-–Vivus’ Qnexa, Arena’s lorcaserin, and Orexigen’s Contrave–were rejected for approval by the FDA, and sent back for further studies. Also in 2010, the then-marketed antiobesity drug sibutramine (Abbott’s Meridia) was withdrawn from the market at the FDA’s request. All of these agents target the central nervous system (CNS).

Concern about long-term safety was the major consideration in the rejection of the NDAs for Qnexa, lorcaserin, and Contrave, and safety issues were also the reason for the withdrawal of sibutramine. That left only one anti-obesity drug approved by the FDA for long term use– orlistat (Roche’s Xenical), with no new drugs In sight. The outlook for obesity drugs was gloomy indeed, and many commentators pronounced the obesity drug field “dead”.

However, as of May 2012, after the further studies prescribed by the FDA in 2010, two of the obesity drug Class of 2010–Qnexa and lorcaserin–had received positive votes by the FDA’s Endocrinologic and Metabolic Drugs Advisory Committee, and had been awaiting final FDA action later this year. Now lorcaserin has been approved. Qnexa is scheduled for an FDA decision by July 17, 2012.

The approval of lorcaserin–especially if Qnexa is also approved–is expected to lift what we have called “the pall of gloom” from the antiobesity drug market. Development of early-stage antiobesity drugs at larger companies that had been put on hold may proceed again, young companies in the field may find it easier to raise capital, and Big Pharma dealmakers may have renewed interest in anitobesity drugs. According to a Jun 28, 2012 article on Bloomberg.com, Big Pharma dealmaking interest has already been aroused.

Limitations of lorcaserin

Despite the excitement over the approval of lorcaserin, the drug has severe limitations.

As we outlined in our September 23, 2010 article on this blog, lorcaserin is a selective serotonin receptor agonist, which is specific for the 5-HT2C serotonin receptor. This contrasts with the nonselective serotonin reuptake inhibitor and serotonin-releasing agents, fenfluramine and dexfenfluramine, which are notorious for their association with heart valve abnormalities.

Lorcaserin was designed to be a more selective agent that works by a similar mechanism to dexfenfluramine or fenfluramine. The anorectic effect of fenfluramine/dexfenfluramine is due to their activity on 5-HT2C, but the adverse effects of these agents appears to be due to their activity on 5-HT2B. Therefore, lorcaserin is expected to be a safer agent that fenfluramine/dexfenfluramine.

However, like fenfluramine and dexfenfluramine, the efficacy of lorcaserin appears to be minimal. Pivotal Phase 3 clinical trials showed an average weight loss of 5.8% among subjects taking lorcaserin, as compared to 2.5% for the placebo group.

A Phase 3 clinical trial published in the New England Journal of Medicine (NEJM) in July 2010 showed that the drug caused significant weight loss and improved maintenance of weight loss as compared to placebo,  in a generally healthy obese population. Lorcaserin also improved values for such biomarkers as lipid levels, insulin resistance, inflammatory markers and blood pressure.

In its July 15, 2010, meeting, the FDA’s Endocrinologic and Metabolic Drugs Advisory Committee noted that lorcaserin, although its efficacy was not great, met FDA efficacy criteria for approvable antiobesity drugs. However, some panelists thought that in populations containing more patients with comorbidities (e.g., diabetes, cardiovascular disease) there might be a lesser degree of efficacy and/or additional safety issues than in populations of generally healthy obese individuals.

However, since that time the results of additional Phase 3 clinical studies in obese individuals with comorbidities, especially type 2 diabetes, have been published. Efficacy results in type 2 diabetics were similar to those seen in obese, nondiabetic individuals. Nevertheless, the efficacy of locaserin remains minimal.

According to the Bloomberg article, analysts believe that insurers will probably not cover lorcaserin due to its low efficacy. However, at a cost of $4 a day for twice-daily therapy with lorcaserin, sales may still reach $2 billion by 2020.

Qnexa, as we discussed in our August 4, 2010 blog article, appears to have a higher efficacy than loracaserin. In the more recent 56-week EQUIP Study of Qnexa in severely obesity individuals (published in February 2012), average weight loss for patients on Qnexa who completed the study was 14.4% and 6.7% with top dose Qnexa and low dose Qnexa, respectively, compared to 2.1% in the placebo group. However, whether Qnexa will be approved awaits the FDA decision by July 12, 2012.

Conclusions

The approval of lorcaserin signals new life for antiobesity drug discovery and development, and the marketing of antiobesity agents. This includes approaches that work by increasing energy expenditure, rather than the usual approaches of decreasing appetite by targeting the CNS. We discussed some of these approaches in our May 23, 2012 article on this blog.

The need for antiobesity agents is great, and with the fast accelerating incidence of obesity and its complications, the need is also accelerating. Moreover, our understanding of the pathogenesis of obesity is limited. Thus both continuing basic research and development of agents with novel mechanisms are sorely needed.

__________________________________________

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.

28 June 2012

Cancer immunotherapy: The star of the 2012 ASCO Annual Meeting

By |2012-06-28T00:00:00+00:00June 28, 2012|Biomarkers, Cancer, Drug Development, Immunology, Monoclonal Antibodies, Personalized Medicine, Strategy and Consulting, Translational Medicine|

 


The American Society of Clinical Oncology (ASCO) held its 2012 Annual Meeting on June 1-5, 2012. Arguably the highlight of the meeting was the June 2, 2012 presentation by Bristol-Myers Squibb (BMS) on its Phase 1 immunotherapeutic, anti-PD-1 (BMS-936558). The results of this study were also published ahead of print on June 2, in the online version of the New England Journal of MedicineNature published a “News in Focus” article on the same subject by Nature staff writer Erika Check Hayden in its 6 June issue.

BMS acquired its anti-PD-1 MAb product BMS-936558 (MDX-1106) via its 2009 acquisition of Medarex. This is the same way in which BMS acquired its now-marketed immunotherapy, ipilimumab (Yervoy), which was approved by the FDA in March 2011. Both BMS-936558 and ipilimumab are monoclonal antibodies (MAbs). Ono Pharmaceuticals has been a partner in the development of anti-PD-1 MAb since its original collaboration with Medarex; Ono retains the right to exclusively develop and market the agent (which is also designated as ONO-4538) in Japan, Korea and Taiwan.

PD-1 (“programmed cell death-1”) is a receptor on the surface of activated T lymphocytes of the immune system. PD-1 is a member of the CD28/CTLA4 family of T cell regulators. Like CTLA4, the target of ipilimumab, PD-1 is a negative regulator of T-cell receptor signals. When a protein on the surface of some tumor cells, known as PD-1 ligand (PD-L1), binds to PD-1 on T cells that recognize antigens on these tumors cells, this results in the blockage of the ability of the T cells to carry out an anti-tumor immune response. Anti-PD-1 MAb binds to PD-1 on T cells, thus preventing PD-L1 on tumor cells from binding to the PD-1 and initiating an inhibitory signal. Anti-tumor T cells are then free to initiate immune responses against the tumor cells. This mechanism of action is completely analogous to that of ipilimumab, which binds to CTLA4 and thus prevents negative signaling from that molecule.

Phase 1 clinical study of Medarex/BMS’s anti-PD-1

The Phase 1 clinical study was carried out by a multi-institution team of investigators, led by Suzanne L. Topalian, M.D. (Johns Hopkins University School of Medicine, Baltimore, MD.) The researchers enrolled patients with advanced melanoma, non-small-cell lung cancer (NSCLC), prostate cancer, renal cell cancer (RCC), or colorectal cancer. Patients received anti-PD-1 at a dose between 0.1 and 10.0 milligrams per kilogram of body weight every two weeks. Tumor response was determined after each 8-week treatment cycle. Patients received up to 12 cycles of treatment until either unacceptable adverse events, disease progression or a complete response occurred. A total of 296 patients received treatment through February 24, 2012.

Among the 236 patients in whom tumor responses could be evaluated, objective responses were observed in patients with NSCLC, melanoma, or RCC. Cumulative response rates (among patients treated with all doses of anti-PD-1) were 18% among patients with NSCLC, 28% among patients with melanoma, and 27% among patients with RCC.  These responses were durable–20 of 31 responses lasted 1 year or more in patients with 1 year or more of follow-up. Anti–PD-1 produced objective responses in approximately one in four to one in five patients with NSCLC, melanoma, or RCC.

In addition to patients with objective responses, other patients treated with anti-PD-1 exhibited stable disease lasting 24 weeks or more–5 patients (7%) with NSCLC, 6 patients (6%) with melanoma, and 9 patients (27%) with RCC.

Significant drug-related adverse effects were seen in 11% of the patients, including three deaths due to pulmonary toxicity. In most cases, adverse effects were reversible, and the observed adverse-event profile does not appear to preclude the use of the drug. A maximum tolerated dose was not reached in this study.

The exciting finding of this study is that anti-PD-1 produced durable responses not only in melanoma and RCC (the two types of cancer that are deemed to be “immunogenic”), but also in NSCLC, a much more common cancer that kills more people per year than any other cancer. Moreover, response rates with anti-PD-1 were much higher that those achieved with the other recently approved immunotherapeutics. In the Phase 3 clinical trial of ipilimumab that led to its approval, this drug gave response rates of 11% in melanoma patients. The other recently approved immunotherapeutic, the prostate cancer-specific dendritic cell vaccine Sipuleucel-T (Dendreon’s Provenge, APC8015), gives very low response rates and no complete responses. According to Antoni Ribas (Jonsson Comp­rehensive Cancer Center, University of California, Los Angeles CA) as quoted Ms. Hayden’s Nature “News in Focus” review, if an immunotherapy “breaks the 10% ceiling” as did anti-PD-1, it becomes “even more important and clinically relevant”.

Despite the exciting efficacy results with anti-PD-1, and despite the fact that it was deemed that the adverse-event profile did not appear to preclude the use of the drug, researchers would still like to get away from the serious adverse effects (including three deaths) seen with anti-PD-1. As with other immunotherapeutics (e.g., ipilimumab), researchers hypothesize that anti-PD-1’s serious adverse effects were due to autoimmune responses.

Phase 1 clinical study of Medarex/BMS’ anti-PD-L1

A potential way of achieving similar efficacy to anti-PD-1 with an improved safety profile is provided by another Phase 1 immunotherapeutic,  anti-PD-L1. Anti-PD-L1 MAb drugs are being developed by Medarex/BMS, Roche/Genentech, and other companies. As mentioned earlier, PD-L1 is the binding partner of PD-1 that is expressed on some tumor cells. As quoted in the Nature “News in Focus” review, Ira Mellman (vice-president of research oncology at Genentech), believes that anti-PD-L1 might have fewer adverse effects than anti-PD-1. That is because anti-PD-L1 would target tumor cells while leaving T cells free to participate in immune networks that work to prevent autoimmune reactions.

The results of a Phase 1 clinical study of BMS/Medarex’ anti-PD-L1 (also known as MDX-1105) were also published ahead of print in the online version of the New England Journal of Medicine on June 2, 2012; this was a “companion study” to the Phase 1 study of anti-PD-1. This study was also carried out by a multi-institution team of investigators, led by Julie R. Brahmer, M.D. (Johns Hopkins University School of Medicine, Baltimore, MD.); Dr. Topalian, among other investigators on the anti-PD-1 trial, also participated in the study.

This Phase 1 trial was a dose escalation study that was carried out via a similar protocol to the anti-PD-1 trial discussed earlier. As of February 24, 2012, a total of 207 patients — 75 with NSCLC, 55 with melanoma, 18 with colorectal cancer, 17 with RCC, 17 with ovarian cancer, 14 with pancreatic cancer, 7 with gastric cancer, and 4 with breast cancer — had received anti–PD-L1 antibody, for a median duration of 12 weeks. Among patients with an evaluable response, an objective response (i.e., a complete or partial response) was seen in 17% of patients with melanoma, 12% of patients with RCC, 10% of patients with NSCLC, and 6% of patients with ovarian cancer. Responses lasted for 1 year or more in 8 of 16 patients with at least 1 year of follow-up. Prolonged disease stabilization was seen in 12-41% of patients with advanced cancers, including NSCLC, melanoma, and RCC.

Significant drug-related adverse effects were seen in 9% of patients.

Although the two agents were not compared directly in a randomized trial, the frequency of objective responses for anti–PD-L1 MAb appears to be somewhat lower than that observed for anti–PD-1 MAb in initial Phase 1 trials; the frequency and severity of significant drug-related adverse events also appears to be lower. However, whether these differences will hold up in Phase 2 and 3 clinical trials remains to be determined. The clinically appropriate dose of anti–PD-L1 will also require further definition later studies. Nevertheless, the Phase 1 trial showed that anti-PD-L1 MAb induced durable tumor regression (objective response rate of 6-17%) and prolonged disease stabilization (rate of 12-41% at 24 weeks) in patients with select advanced cancers, including NSCLC, a tumor type that had been deemed to be “non-immunogenic”. This is essentially the same result that was observed for anti-PD-1MAb.

A predictive biomarker for treatment with anti-PD-1?

As with other modes of cancer therapy, it would be very useful to have mechanism-based predictive biomarkers to identify appropriate candidates for treatment with anti-PD-1 or anti-PD-L1 immunotherapy. The findings of the Phase 1 anti-PD-1 study suggest that PD-L1 expression in tumors is a candidate biomarker that warrants further evaluation for use in selecting patients for immunotherapy with anti–PD-1 MAb. The researchers found that 36% of patients with PD-L1–positive tumors achieved an objective response, while no patients with PD-L1–negative tumors achieved such a response. These results suggest that PD-L1 expression on the surface of tumor cells in pre-treatment tumor specimens may be associated with an objective response. However, further studies will be necessary to define the role of PD-L1 as a predictive biomarker of response to anti–PD-1 therapy. Similarly, it appears reasonable that tumor expression of PD-L1 may be a predictive biomarker of response to anti-PD-L1 therapy. However, this hypothesis must also be tested in further clinical studies.

Further studies of anti-PD-1 MAb

Two studies of BMS-936558/MDX-1106 anti–PD-1 MAb, both in advanced/metastatic clear-cell RCC, are now recruiting patients. One trial is a Phase 1 biomarker study involving immunologic and tumor marker correlates of efficacy (progression-free survival and tumor response). The other trial is a Phase 2 efficacy (progression-free survival and tumor response) study; this is a dose ranging study that is designed to determine if a dose response exists. Phase 3 studies of BMS-936558/MDX-1106 anti–PD-1 MAb for the treatment of non–small-cell lung cancer, melanoma, and renal-cell cancer are also being planned.

Conclusions

The exciting results of the studies with BMS’ anti-PD-1 and anti-PD-L1 have only been in Phase 1 studies. Thus caution is advisable in interpreting these results, pending the results of further clinical studies. Nevertheless, these results, together with the recent approval of ipilimumab (Medarex/Bristol-Myers Squibb’s Yervoy) and of Sipuleucel-T (Dendreon’s Provenge), indicate that cancer immunotherapy, a field that not so long ago was regarded as an impractical dream, is very much alive and well. In addition to clinical development and approval of immunotherapeutic agents, exciting basic and drug discovery research in this field is ongoing. This was recognized by the awarding of the 2011 Nobel Prize in Physiology or Medicine for research with profound implications for the development of cancer immunotherapies.

The Biopharmconsortium Blog has been covering new developments in cancer immunotherapy since the spring of 2011. Our earlier articles on this subject (with links) are listed in our December 31, 2011 article, entitled “Read the cancer immunotherapy review in the 22 December 2011 issue of Nature!”

Cancer immunotherapy represents one of several “scientifically premature” or “frontier science” areas discussed in this blog that are providing new opportunities for drug discovery and development–for young entrepreneurial biotech start-ups and for more established biotechnology and pharmaceutical companies. Corporate strategists would do well to explore such areas for potential new R&D programs for their companies.

__________________________________________

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.

11 June 2012

Developing resistance-free antibiotics by targeting quorum sensing

By |2018-12-28T23:36:25+00:00June 11, 2012|Chemistry, Drug Development, Drug Discovery, Haberman Associates, Infectious Disease, Strategy and Consulting, Synthetic biology|

 

Quorum sensing synthetic biology project http://bit.ly/LO1ynR

Way back in May 2000, Decision Resources published my short report entitled “New approaches to small-molecule antibacterial drug discovery” as part of its Spectrum Life Sciences series. As might be expected, the report is now out of print.

The report was a brief review of then-novel approaches to antibacterial drug discovery, in the face of the increasing level of antibiotic resistance in pathogenic bacteria. These approaches included genomics and such technologies as high-throughput screening against bacterial-specific targets.

However, the most interesting part of the report was a section on using the study of bacterial physiology to identify targets that are important for the ability of bacteria to cause disease, but are not essential for bacterial proliferation or survival. The hypothesis behind these studies was that it might be possible to develop compounds that prevent these bacteria from causing disease, without selecting for resistant strains of the bacteria.

Antibiotics typically kill or prevent proliferation of bacteria by targeting biomolecules involved in such essential processes as cell wall synthesis, DNA proliferation, or protein synthesis. Treating large populations of bacteria with such agents inevitably selects for a few resistant mutant cells. These proliferate, mutate further, and give rise to antibiotic resistant populations. However, if a therapeutic targets a nonessential pathway that is involved in pathogenesis, resistant populations might not be selected for. That was the hypothesis.

This field of bacterial physiology for drug discovery focused on two related areas–virulence factors and quorum sensing. Virulence factors are not expressed by a strain of pathogenic bacteria in vitro, but are expressed only when the bacteria infect a host. Once expressed, they enable the bacteria to colonize the host and cause disease. Examples of such virulence factors include secretion systems that deliver bacterial effector proteins into host cells. These effector proteins may, for example, kill host cells, inhibit cytokine production or phagocytosis, or may mediate bacterial entry into the host cells.

Quorum sensing is a system by which certain bacteria can monitor their own population density. They accomplish this by secreting specific autoinducer molecules. When the concentration of an autoinducer reaches a critical threshold value (as the result of an increase in bacterial population density), it triggers specific response systems, causing the induction of sets of genes that are only expressed at high population density.

For example, many gram-negative bacteria (e.g., Pseudomonas aeruginosa, Vibrio cholerae, and Escherichia coli) use specific acyl homoserine lactones (AHSLs) as their autoinducers. P. aeruginosa has two quorum sensing systems that use the AHSL autoinducers butyrylhomoserinelactone and 3-oxododecanoylhomoserinelactone, respectively. These systems (operating via specific receptors for the auotoinducers and interacting with each other) control the induction of several genes, some of which are virulence factors. Some of these genes enable the bacteria, when they are at sufficient density, to form biofilms (slimy mats of bacteria and polysaccharide matrix).

P. aeruginosa is an opportunistic pathogen, causing infection in the lungs of people with cystic fibrosis, burn patients, and other hospitalized patients. These infections cause death in over 80% of cystic fibrosis patients. The ability to form biofilms renders the bacteria resistant to antibiotics and to the patient’s own immune system.

Other gram-negative bacteria that form biofilms have been implicated in dental caries, peridontitis, osteomyelitis, and numerous nosocomial infections. Bacterial biofilms can also form on the surface of implanted medical devices, such as catheters and mechanical heart valves, and cause device-related infections.

The gram-positive human pathogen Staphylococcus aureus also has a quorum sensing system. However, it does not use an AHSL as an autoinducer. The S. aureus autoinducers are peptides that contain an unusual thiolactone structure (i.e., a thol ester-linked cyclic structure). The S. aureus quorum sensing system controls the synthesis of virulence factors responsible for the pathogenicity of this organism in vivo. Although specific peptides induce virulence factors in a given strain of S. aureus, there are other specific peptides that inhibit the induction of virulence in strains of the organism other than the one secreting the inhibitory peptides. That finding suggested that researchers should be able to develop specific agents to shut down S. aureus pathogenesis by targeting the quorum sensing system.

Interestingly, quorum sensing-based systems have been used in projects for the International Genetically Engineered Machine (iGEM) competition, an annual undergraduate synthetic biology competition. See the figure above, which was taken from the 2009 Chiba University (Japan) iGEM project.  [http://2009.igem.org/Team:Chiba/Project/Signaling-system]

Quorum Sciences and Vertex Pharmaceuticals’ research on quorum sensing

At the time of the writing and publication of our antibacterial drug discovery report, there was a company, Quorum Sciences (Iowa City, IA) that had been established to commercialize the findings of leading researchers on bacterial quorum sensing. As the result of two successive acquisitions in 2000 and 2001, Quorum Sciences passed into the hands of Vertex Pharmaceuticals (Cambridge, MA). In 2006, Vertex researchers and their academic collaborators published a report on the discovery of novel specific inhibitors of the P. aeruginosa quorum sensing system. The last author of this report was quorum sensing pioneer E. Peter Greenberg, formerly of the University of Iowa and chief scientific officer at Quorum Sciences, and from 2005 to the present at the University of Washington School of Medicine. The compounds identified in the 2006 report, discovered via high-throughput screening of a diverse 200,000-compound chemical library, resembled the natural AHSL that binds to the P. aeruginosa quorum sensing receptor LasR. (LasR is a transcription factor that when bound to its specific AHSL, mediates the expression of a set of downstream genes, including those that encode virulence factors.) The researchers concluded that the novel quorum sensing inhibitors might be useful chemical tools, but not drug leads.

In 2010, other academic researchers published a report on the discovery of novel antagonists and agonists of the P. aeruginosa quorum sensing receptor LasR, which were of lower molecular weight and otherwise structurally distinct from the natural P. aeruginosa AHSL. However, these compounds were still deemed to be scaffolds for chemical tools, not drug leads. Nevertheless, the researchers speculated that the compounds “could, with further development, provide a pathway for the design of novel antivirulence agents”. Other researchers are continuing studies aimed at discovery of quorum sensing receptor antagonists, whether synthetic organic molecules or natural products. These involve studies with quorum sensing systems of both gram-positive and gram-negative bacteria.

The 2006 report appears to be the last Vertex publication on quorum sensing. However, Vertex continues to conduct research on antibacterial agents. And the company has a facility in the University of Iowa BioVentures Center (Coralville, IA),  which is a continuation of the old Quorum Sciences Iowa facility. As of 2009, Vertex’s Iowa-based team consisted of seven full-time scientists, working on development of antibacterials, and agents to treat hepatitis C and cystic fibrosis, among other areas. The Iowa group participated in the development of Vertex’ now-marketed anti-hepatitis C virus (HCV) agent Incivek (telaprevir).

The May 2012 article “Freezing Time” in The Scientist, and discovery of novel quorum sensing inhibitors

The May 2012 issue of The Scientist contains an article entitled “Freezing Time”, by Vern L Schramm, Ph.D. (Albert Einstein College of Medicine (Bronx, NY). The article focused on design of “transition state analogues”, i.e., compounds with a chemical structure that resembles the transition state of a substrate in an enzyme-catalyzed reaction. Transition state analogs usually act as enzyme inhibitors by blocking the enzyme’s active site. They are exquisitely potent and specific inhibitors, which act at extremely small doses. This makes these compounds potentially attractive as drugs.

A transition state analogue inhibitor that was designed by Dr. Schramm and his colleagues in the early 2000s as an early proof-of-concept molecule is immucillin-H, or forodesine. This is a transition-state analog inhibitor of purine nucleoside phosphorylase.  Forodesine is being developed by BioCryst Pharmaceuticals for treatment of relapsed B-cell chronic lymphocytic leukemia, and the results of a Phase 2 trial were published in 2010.

As described in Dr. Schramm’s May 2012 article, his laboratory has been applying their transition-state analogue technology to the field of quorum sensing in bacteria. Instead of targeting the recognition of AHSLs by quorum sensing receptors such as LasR, the researchers targeted the key enzyme in the AHSL biosynthesis pathway in gram-negative bacteria, known as 5′-methylthioadenosine nucleosidase (MTAN). The biosynthetic pathway for the production of AHSLs, including the key role of MTAN, had been elucidated by Dr. Greenberg and his colleagues in the late 1990s.

Dr. Schramm and his colleagues published the results of studies of three transition state analogues that potently inhibited MTANs of gram-negative bacteria. For example, they inhibited the Vibrio cholerae MTAN with dissociation constants of 73, 70, and 208 pM, respectively. They inhibited MTAN in cell of a virulent strain of V. cholerae with IC50 values of 27, 31, and 6 nM respectively, disrupting autoinducer production in a dose-dependent manner without affecting bacterial growth. The compounds were also potent inhibitors of autoinducer production in an enterohemorrhagic strain of Escherichia coli. The transition-state analogues did not inhibit growth in either V. cholerae or E. coli, but one such compound reduced biofilm production by 18% in E. coli and 71% in V. cholerae.

Moreover, the MTAN inhibitors did not appear to select for bacterial resistance in vitro. When V. cholerae bacteria were grown for 26 generations in the presence of a large excess of MTAN inhibitors, subsequent generations of these bacteria were equally sensitive to inhibition by these compounds as bacteria that had not been previously exposed to the inhibitors. These results are consistent with the hypothesis that agents that inhibit targets that are important in the ability of bacteria to cause disease, but are not essential for bacterial proliferation or survival might not select for drug resistance.

As Dr. Schramm said in the May 2012 article in The Scientist, it remains to be seen whether the MTAN-targeting transition-state analogs developed in his laboratory can translate into novel antibiotics that do not select for resistant pathogens. As of March 2009, Dr. Schramm’s team had developed over 20 potent MTAN inhibitors, which will be specific for bacteria and should have no effect on human metabolism. These compounds have been licensed to Pico Pharmaceuticals (Melbourne, Australia), which plans to develop and initiate clinical trials. Dr. Schramm is a Pico Pharmaceuticals co-founder and chairman of its scientific advisory board. Pico claims that one of its quorum sensing inhibitors, designated as PC0208, has demonstrated proof-of-concept in preclinical studies, and now has “pre-IND” status.

Lessons from these studies

Dr. Schramm’s discovery of novel quorum sensing inhibitors was made possible by a strategy that involved a combination of biology-driven drug discovery and sophisticated chemistry technology. The biology-driven drug discovery strategy involved a combination of 1. Building on the quorum sensing studies of Dr. Greenberg and others, and adopting the strategy, as reviewed in our 2000 Spectrum report, of targeting the quorum sensing system in order to discover agents that would have the possibility of not triggering resistance, and 2. Targeting a critical, bacterial-specific pathway enzyme that is upstream of the recognition of AHSLs by quorum sensing receptors (the usual target of most researchers in this area). This enzyme, MTAN, has a key role in the biosynthesis of AHSLs.

The sophisticated chemical technology employed by Dr. Schramm and his colleagues was of course the transition state analogue technology developed in his own laboratory. Combined with the biology-driven strategy described in the last paragraph, Dr. Schramm’s approach has succeeded in the discovery of compounds that are potential drug candidates, while approaches based on high-throughput screening for AHSL antagonists have so far failed to produce any such compounds. Dr. Scharamm’s laboratory has also obtained evidence that treatment with their compounds should not result in the selection of resistant strains of pathogenic bacteria.

It is possible that other chemistry approaches might be successfully employed to discover quorum sensing inhibitors, both for gram-negative bacteria and gram-positive organisms such as S. aureus.

As we have discussed in numerous articles on this blog, biology-driven drug discovery strategies, often coupled with innovative approaches to chemistry (in the case of small-molecule drug discovery) are applicable to very many different targets involved in a whole range of human diseases. (Biology-driven drug discovery has also been central to discovery and development of many successful large-molecule drugs.) The quorum sensing case study in this article is a simple, understandable, and elegant example of such a strategy.

In addition to the scientific, clinical, and medical aspects of antibacterial drug discovery, the other major issue is the business of antibacterial discovery and development. The economics of drug discovery and development have shifted pharmaceutical industry investment away from the development of drugs targeting short course therapies for acute diseases (such as antibacterials) and towards long-term treatment of chronic conditions.  At the same time, discovery of novel antibacterials has gotten more difficult. As a result, during the 2000-2010 period, such companies as Wyeth, Aventis, Eli Lilly, GlaxoSmithKline, Bristol-Myers Squibb, Abbott Laboratories, Proctor & Gamble, and Merck have either deprioritized anti-bacterial R&D or left the field altogether. Meanwhile, antibiotic resistance, which was a problem in 2000, has become an even greater problem in 2012, in some cases reaching crisis proportions [e.g, methicillin resistant S. aureus (MRSA) that is also resistant to the drug of last resort, vancomycin].

As a result of these economic, scientific, and medical challenges, a €223.7 consortium of five pharmaceutical companies and leading academics, called NewDrugs4BagBugs (ND4BB) was launched in Europe in May 2012. The program is envisioned to involve a three-stage approach – to improve the understanding of antimicrobial resistance, to design and implement efficient clinical trials, and finally, to take novel drug candidates through clinical development.

And at least one venture capitalist has observed that biotechs that specialize in antibacterial drug development (as well as those that specialize in other areas that have been deemphasized by Big Pharmas) have provided “contrarian opportunities” in biotech venture. According to a June 2 2012 article by Bruce Booth of Atlas Venture published in Forbes, what has been deprioritized by some (or several) Big Pharmas, are likely be re-prioritized by others several years later. Such antibacterial drug developers as Calixa, Cerexa, Novexel, Neutec, Paratek, Pennisula, Protez, and Vicuron have produced some of the best returns in biotech venture capital from merger/acquisition exits. These biotechs included companies that were built around compounds outlicensed from Big Pharma, and others that conducted new research on novel targets, especially for MRSA and other resistant bacteria.  By taking advantage of a strategic depriorization in Pharma, these biotechs and their venture backers were able to create considerable value in the past decade out of antibacterial drug development.

Meanwhile, antibiotic specialist Cubist Pharmaceuticals (Lexington, MA) remains an independent, and profitable, biotech company that is continuing to conduct R&D, including on discovery and development of agents to treat pathogens that are resistant to current antibiotics. It has expanded into development and marketing of peripheral mu-opioid receptor antagonists (including via acquisition of Adolor in 2011), and has recently expanded its R&D facilities.

Can Pico Pharmaceuticals (which has oncology programs in addition to antibacterials) experience similar success?

__________________________________________

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 contact us by phone or e-mail. We also welcome your comments on this or any other article on this blog.

Go to Top