none | Boulder Peptide Symposium

September 15-18, 2025

LIVE, In Person at the St. Julien Hotel in Boulder, Colorado
The only conference focused solely on the pharmaceutical development of peptide therapeutics.

All posts in none

Reappraisal of GIP Pharmacology for Metabolic Diseases.

Source:

Trends Mol Med. 2016 May;22(5):359-76. doi: 10.1016/j.molmed.2016.03.005. Epub 2016 Mar 30.

Abstract

Glucagon-like peptide-1 (GLP-1) analogs are considered the best current medicines for type 2 diabetes (T2D) and obesity due to their actions in lowering blood glucose and body weight. Despite similarities to GLP-1, glucose-dependent insulinotropic polypeptide (GIP) has not been extensively pursued as a medical treatment for T2D. This is largely based on observations of diminished responses of GIP to lower blood glucose in select patients, as well as evidence from rodent knockout models implying that GIP promotes obesity. These findings have prompted the belief in some, that inhibiting GIP action might be beneficial for metabolic diseases. However, a growing body of new evidence - including data based on refined genetically modified models and improved pharmacological agents - suggests a paradigm shift on how the GIP system should be manipulated for metabolic benefits.

Finan B1, Müller TD1, Clemmensen C1, Perez-Tilve D2, DiMarchi RD3, Tschöp MH4.

Author information

  • 1Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
  • 2Metabolic Diseases Institute, Division of Endocrinology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
  • 3Department of Chemistry, Indiana University, Bloomington, IN, USA.
  • 4Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany. Electronic address: matthias.tschoep@helmholtz-muenchen.de.
Oral Administration of Peptide-Based Drugs: Beyond Lipinski’s Rule.

Source:

ChemMedChem. 2016 Sep 6. doi: 10.1002/cmdc.201600288. [Epub ahead of print]

Abstract

The use of peptides in therapy presents several limitations, from physicochemical characteristics to inadequate pharmacokinetic profiles for oral absorption. As peptides are gaining importance in the therapeutic arsenal, there is an increasing need to rationalize the main characteristics of this compound class in the market. Therefore, we performed an extensive analysis of all known peptide drugs and clinical candidates based on their peptide features, physicochemical and structural properties, and correlated these with their administration route and therapeutic classes. Peptide drugs are widely distributed across drug and pharmacological space, covering several therapeutic areas with structural diversity and complexity, distributed between groups of cyclic and linear compounds. Although structural and physicochemical properties are clear within these groups, we counter the consensus that cyclic peptides have better oral availability than linear peptides, as most of the orally administrated peptides have linear structures. This study and review furnishes information that could support peptide drug design, with a new cutoff of known descriptors that go beyond the Rule of Five.

Santos GB1, Ganesan A2, Emery FS3.

Author information

  • 1School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Cafe s/n, Cidade Universitaria, Ribeirao Preto, SP, Brazil.
  • 2School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
  • 3School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Cafe s/n, Cidade Universitaria, Ribeirao Preto, SP, Brazil. flavioemery@fcfrp.usp.br.
Meet the 2016 Young Investigator Award Recipient

The Boulder Peptide Society is pleased to announce that Dr. Albert Bowers, at the University of North Carolina, has received the Young Investigator Award for 2016.  The Young Investigator Award was established to support promising peptide scientists during the pre-tenure period. 

Albert Bowers is Assistant Professor in the Division of Chemical Biology & Medicinal Chemistry at the Eshelman School of Pharmacy of the University of North Carolina at Chapel Hill. Albert received his PhD in organic chemistry (synthetic methods) from the University of Illinois at Chicago under the direction of Dr. David Crich. He carried out postdoctoral research in total synthesis at Colorado State University under the direction of Robert M. Williams, then moved to an NIH sponsored fellowship at Harvard Medical School to continue postdoctoral research in biosynthesis under the direction of Christopher T. Walsh. In addition to his appointment in pharmacy at UNC, Albert is a member of the UNC Lineberger Comprehensive Cancer Center and affiliate member of the Center for Integrative Chemical Biology and Drug Discovery.

Albert Bowers has published 30 manuscripts including 11 in the Journal of the American Chemical Society and 12 since becoming an independent investigator. Albert has received the prestigious Beckman Young Investigator (BYI) Award from the Arnold and Mabel Beckman Foundation. Research in the Bowers lab focuses on chemistry and biotechnology for creating libraries of natural product-like cyclic peptides, in particular by using enzymes from RiPPs (ribosomally translated and posttranslationally modified peptide natural product) pathways. Using an innovative combination of total chemical synthesis, native chemical ligation, and enzymology, his lab was the first to characterize the unique pyridine synthases from the thiopeptide pathways, and answering a long standing question in the biosynthesis of these compounds. Building upon this work, they developed a solid-phase route to thiopeptides with improved activity and solubility. The Bowers lab has also structurally and biochemically characterized a unique E1-like enzyme from the biosynthesis of the pantocin, peptide-derived natural products. By adapting these unique enzymes to various formats of peptide libraries, the Bowers lab aims to develop a combinatorial biosynthesis platform for the discovery of peptide natural product-like inhibitors.

In recognition of his professional and scientific achievements, the Scientific Advisory Board of the Boulder Peptide Society is pleased to present the award to Albert Bowers. Dr. Bowers will present an oral presentation on his research and formally accept the award at the fall Boulder Peptide Symposium Sept 26-29, 2016 in Boulder, Colorado.

Chemical Synthesis of Human Insulin-Like Peptide-6.

Source:

Chemistry. 2016 Jun 3. doi: 10.1002/chem.201601410. [Epub ahead of print]

Abstract

Human insulin-like peptide-6 (INSL-6) belongs to the insulin superfamily and shares the distinctive disulfide bond configuration of human insulin. In this report we present the first chemical synthesis of INSL-6 utilizing fluorenylmethyloxycarbonyl-based (Fmoc) solid-phase peptide chemistry and regioselective disulfide bond construction protocols. Due to the presence of an oxidation-sensitive tryptophan residue, two new orthogonal synthetic methodologies were developed. The first method involved the identification of an additive to suppress the oxidation of tryptophan during iodine-mediated S-acetamidomethyl (Acm) deprotection and the second utilized iodine-free, sulfoxide-directed disulfide bond formation. The methodologies presented here offer an efficient synthetic route to INSL-6 and will further improve synthetic access to other multiple-disulfide-containing peptides with oxidation-sensitive residues.

 

Wu F1, Mayer JP1, Zaykov AN1, Zhang F1, Liu F2, DiMarchi RD3.

Author information

  • 1Department of Chemistry, Indiana University Bloomington, 800 E. Kirkwood Avenue, Bloomington, Indiana, 47405, USA.
  • 2Novo Nordisk Research Center Indianapolis, 5225 Exploration Drive, Indianapolis, Indiana, 46241, USA. falx@novonordisk.com.
  • 3Department of Chemistry, Indiana University Bloomington, 800 E. Kirkwood Avenue, Bloomington, Indiana, 47405, USA. rdimarch@indiana.edu.
The Extracellular Surface of the GLP-1 Receptor Is a Molecular Trigger for Biased Agonism.

Source:

Cell. 2016 Jun 16;165(7):1632-43. doi: 10.1016/j.cell.2016.05.023.

Abstract

Ligand-directed signal bias offers opportunities for sculpting molecular events, with the promise of better, safer therapeutics. Critical to the exploitation of signal bias is an understanding of the molecular events coupling ligand binding to intracellular signaling. Activation of class B G protein-coupled receptors is driven by interaction of the peptide N terminus with the receptor core. To understand how this drives signaling, we have used advanced analytical methods that enable separation of effects on pathway-specific signaling from those that modify agonist affinity and mapped the functional consequence of receptor modification onto three-dimensional models of a receptor-ligand complex. This yields molecular insights into the initiation of receptor activation and the mechanistic basis for biased agonism. Our data reveal that peptide agonists can engage different elements of the receptor extracellular face to achieve effector coupling and biased signaling providing a foundation for rational design of biased agonists.

 

Wootten D1, Reynolds CA2, Smith KJ2, Mobarec JC2, Koole C3, Savage EE4, Pabreja K4, Simms J5, Sridhar R4, Furness SG4, Liu M6, Thompson PE6, Miller LJ7, Christopoulos A4, Sexton PM8.

Author information

  • 1Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. Electronic address: denise.wootten@monash.edu.
  • 2School of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK.
  • 3Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
  • 4Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
  • 5School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK.
  • 6Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
  • 7Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA.
  • 8Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. Electronic address: patrick.sexton@monash.edu.
Π-Clamp-mediated cysteine conjugation.

Source:

Nat Chem. 2016 Feb;8(2):120-8. doi: 10.1038/nchem.2413. Epub 2015 Dec 21.

Abstract

Site-selective functionalization of complex molecules is one of the most significant challenges in chemistry. Typically, protecting groups or catalysts must be used to enable the selective modification of one site among many that are similarly reactive, and general strategies that selectively tune the local chemical environment around a target site are rare. Here, we show a four-amino-acid sequence (Phe-Cys-Pro-Phe), which we call the 'π-clamp', that tunes the reactivity of its cysteine thiol for site-selective conjugation with perfluoroaromatic reagents. We use the π-clamp to selectively modify one cysteine site in proteins containing multiple endogenous cysteine residues. These examples include antibodies and cysteine-based enzymes that would be difficult to modify selectively using standard cysteine-based methods. Antibodies modified using the π-clamp retained binding affinity to their targets, enabling the synthesis of site-specific antibody-drug conjugates for selective killing of HER2-positive breast cancer cells. The π-clamp is an unexpected approach to mediate site-selective chemistry and provides new avenues to modify biomolecules for research and therapeutics.

 

Zhang C1, Welborn M1, Zhu T1, Yang NJ2, Santos MS2, Van Voorhis T1, Pentelute BL1.

Author information

  • 1Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA.
  • 2Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA.
Efficacy and Safety of LixiLan, a Titratable Fixed-Ratio Combination of Lixisenatide and Insulin Glargine, Versus Insulin Glargine in Type 2 Diabetes Inadequately Controlled on Metformin Monotherapy: The LixiLan Proof-of-Concept Randomized Trial

Source:

Diabetes Care. 2016 Jun 9. pii: dc160046. [Epub ahead of print]

Abstract

OBJECTIVE:

This study assessed the efficacy and safety of LixiLan, a fixed-ratio, titratable, combination of 2 units insulin glargine (Gla-100) and 1 μg lixisenatide administered once daily via a single pen, versus Gla-100 in insulin-naïve type 2 diabetes on metformin.

RESEARCH DESIGN AND METHODS:

Participants were randomized to once-daily LixiLan (n = 161) or Gla-100 (n = 162) for 24 weeks, while continuing metformin. LixiLan and Gla-100 were started at 10 units/5 μg and 10 units, respectively, and titrated based on the Gla-100 requirement according to fasting plasma glucose levels. The primary objective was to test noninferiority (upper bound of the 95% CI ≤0.4%) of LixiLan in reducing HbA1c; if met, statistical superiority was tested. Secondary objectives included body weight changes, hypoglycemia, and safety.

RESULTS:

Baseline characteristics (mean age 57 years, diabetes duration 6-7 years, BMI 32 kg/m2) were similar between groups. At week 24, mean HbA1c was reduced from 8.0% (64 mmol/mol) at baseline to 6.3% (45 mmol/mol) and 6.5% (48 mmol/mol) with LixiLan and Gla-100, respectively, establishing statistical noninferiority and superiority of LixiLan (least-squared mean [95% CI] difference: -0.17% [-1.9 mmol/mol] (-0.31, -0.04% [-3.4, -0.4 mmol/mol]; P = 0.01). HbA1c <7.0% (<53 mmol/mol) was achieved in 84% and 78% of participants (nonsignificant), respectively. LixiLan improved 2-h postmeal plasma glucose versus Gla-100 (least-squared mean difference: -3.17 mmol/L (-57 mg/dL); P < 0.0001). Body weight was reduced with LixiLan (-1 kg) and increased with Gla-100 (+0.5 kg; P < 0.0001), with no increase in hypoglycemic events (25% in each group). The incidence of nausea (7.5%) and vomiting (2.5%) was low with LixiLan.

CONCLUSIONS:

LixiLan achieved statistically significant reductions to near-normal HbA1c levels with weight loss and no increased hypoglycemic risk, compared with insulin glargine alone, and a low incidence of gastrointestinal adverse events in type 2 diabetes inadequately controlled on metformin.

 

Rosenstock J1, Diamant M2, Aroda VR3, Silvestre L4, Souhami E4, Zhou T5, Perfetti R5, Fonseca V6; LixiLan PoC Study Group.

Author information

  • 1Dallas Diabetes and Endocrine Center at Medical City, Dallas, TX juliorosenstock@dallasdiabetes.com.
  • 2VU University Medical Center, Amsterdam, the Netherlands.
  • 3MedStar Health Research Institute, Hyattsville, MD.
  • 4Sanofi, Paris, France.
  • 5Sanofi, Bridgewater, NJ.
  • 6Tulane University Health Sciences Center, New Orleans, LA.
The development and use of the E75 (HER2 369-377) peptide vaccine

source:

Future Oncol. 2016 Jun;12(11):1321-9. doi: 10.2217/fon-2015-0054. Epub 2016 Apr 5.

Abstract

E75 (nelipepimut-S) is an immunogenic peptide derived from the HER2 protein. When combined with the immunoadjuvant granulocyte-macrophage colony-stimulating factor (GM-CSF), nelipepimut-S has been used as a vaccine that is capable of eliciting a robust anti-HER2 immune response. Early-phase clinical trials that enrolled women with node-positive or high-risk node-negative breast cancer who had been rendered disease free with standard of care therapy but were at risk for recurrence, demonstrated the vaccine to be safe with a suggestion of clinical benefit. Nelipepimut-S is currently being evaluated in a Phase III clinical trial. This article covers the preclinical and clinical development of nelipepimut-S.

 

Clifton GT1, Peoples GE2, Mittendorf EA3.

Author information

  • 1Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX 77030, USA.
  • 2Cancer Vaccine Development Program, Metis Foundation, 600 Navarro Street, San Antonio, TX 78205, USA.
  • 3Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1434, Houston, TX 77030, USA.
Development of a prosaposin-derived therapeutic cyclic peptide that targets ovarian cancer via the tumor microenvironment.

Source:

Sci Transl Med. 2016 Mar 9;8(329):329ra34. doi: 10.1126/scitranslmed.aad5653.

Abstract

The vast majority of ovarian cancer-related deaths are caused by metastatic dissemination of tumor cells, resulting in subsequent organ failure. However, despite our increased understanding of the physiological processes involved in tumor metastasis, there are no clinically approved drugs that have made a major impact in increasing the overall survival of patients with advanced, metastatic ovarian cancer. We identified prosaposin (psap) as a potent inhibitor of tumor metastasis, which acts via stimulation of p53 and the antitumorigenic protein thrombospondin-1 (TSP-1) in bone marrow-derived cells that are recruited to metastatic sites. We report that more than 97% of human serous ovarian tumors tested express CD36, the receptor that mediates the proapoptotic activity of TSP-1. Accordingly, we sought to determine whether a peptide derived from psap would be effective in treating this form of ovarian cancer. To that end, we developed a cyclic peptide with drug-like properties derived from the active sequence in psap. The cyclic psap peptide promoted tumor regression in a patient-derived tumor xenograft model of metastatic ovarian cancer. Thus, we hypothesize that a therapeutic agent based on this psap peptide would have efficacy in treating patients with metastatic ovarian cancer.

π-Clamp-mediated cysteine conjugation.

Source:

Nat Chem. 2016 Feb;8(2):120-8. doi: 10.1038/nchem.2413. Epub 2015 Dec 21.

Abstract

Site-selective functionalization of complex molecules is one of the most significant challenges in chemistry. Typically, protecting groups or catalysts must be used to enable the selective modification of one site among many that are similarly reactive, and general strategies that selectively tune the local chemical environment around a target site are rare. Here, we show a four-amino-acid sequence (Phe-Cys-Pro-Phe), which we call the 'π-clamp', that tunes the reactivity of its cysteine thiol for site-selective conjugation with perfluoroaromatic reagents. We use the π-clamp to selectively modify one cysteine site in proteins containing multiple endogenous cysteine residues. These examples include antibodies and cysteine-based enzymes that would be difficult to modify selectively using standard cysteine-based methods. Antibodies modified using the π-clamp retained binding affinity to their targets, enabling the synthesis of site-specific antibody-drug conjugates for selective killing of HER2-positive breast cancer cells. The π-clamp is an unexpected approach to mediate site-selective chemistry and provides new avenues to modify biomolecules for research and therapeutics.


s2Member®
loading...