News | 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 News

BPF Journal Club – May edition

Disrupting Cancer Signaling with Precision: The Promise of GO-Pep as a Targeted BAG-1 Inhibitor

For our May Journal Club, we review a recently published article in Communications Biology. Özge Tatli1 and colleagues report the development of GO-Pep, a rationally designed peptide that disrupts the interaction between the co-chaperone BAG-1 and the kinase c-Raf—two proteins implicated in promoting cancer cell survival through the MAPK/ERK signaling pathway. Using hydrogen-deuterium exchange mass spectrometry (HDX-MS), the team mapped the critical binding interface between BAG-1 and a 20-amino acid stretch of c-Raf, which they used to construct GO-Pep. This peptide, fused to a cell-penetrating motif, successfully entered cancer cells and displaced c-Raf from its complex with BAG-1, leading to a reduction in downstream MAPK signaling. GO-Pep treatment resulted in increased apoptosis and reduced proliferation in BAG-1-overexpressing cancer cell lines, with minimal cytotoxicity observed in non-cancerous cells—highlighting its therapeutic potential.

This work exemplifies how detailed protein interaction mapping can drive the design of peptides that selectively modulate protein-protein interactions (PPIs), long considered challenging drug targets. GO-Pep’s success in vitro sets the stage for further optimization, including stability enhancement and targeted delivery strategies, to realize its translational potential in cancer therapy. For the peptide community at BPF, GO-Pep is a compelling case of structure-guided PPI disruption that aligns with broader efforts to expand the druggable proteome using peptide-based modalities.

1Link to open access article: https://doi.org/10.1038/s42003-024-07419-4

Tatlı, Ö., Eyüpoğlu, E., Er, Y., Aytekin, E., Yılmaz, B., Cingöz, A., Akgül, C., & Doğanlar, O. (2025). A BAG-1-inhibitory peptide, GO-Pep, suppresses c-Raf activity in cancer. Communications Biology, 8, Article 235.

Kevin McCowen
Treasurer Boulder Peptide Foundation, Member Board of Directors
President MAC-Lab
linkedin.com/in/kevin-mccowen-a40b8030

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 

BPF Journal Club – April edition

Hit-to-Lead: Turning Hits into Promising Drug Candidates

This month’s article, authored by a group of researchers from multiple institutions collaborating with the European Federation for Medicinal Chemistry and Chemical Biology (EFMC) (https://doi.org/10.1002/cmdc.202400931), explores the critical transition from initial drug discovery hits to optimized lead compounds, highlighting key challenges and strategies in this pivotal phase of drug development.

The Hit-to-Lead (H2L) stage is a critical phase in drug discovery, where initial hits identified through high-throughput screening, virtual screening, or small molecule/fragment-based approaches undergo systematic refinement to yield lead compounds with improved pharmacological and physicochemical properties. This process marks a key decision-making juncture, ensuring that only the most promising chemical series advance into lead optimization while mitigating liabilities related to potency, selectivity, pharmacokinetics, and toxicity. While these principles have been extensively applied to small molecules, they are equally relevant in peptide drug discovery, where additional challenges—such as proteolytic stability, permeability, and half-life extension—must be addressed. Strategies like backbone modifications, non-natural amino acid incorporation, and macrocyclization help optimize peptide leads, paralleling the rational design approaches used for small molecules.

A fundamental aspect of H2L is the prioritization and selection of chemical matter based on a comprehensive assessment. While potency is an essential parameter, it must be balanced with selectivity to minimize off-target interactions that could lead to adverse effects. Physicochemical properties such as lipophilicity, solubility, metabolic stability, and permeability play a crucial role in drug-likeness and should be optimized early. Excessive lipophilicity often leads to poor metabolic stability, rapid clearance, and off-target binding, whereas low permeability may result in poor bioavailability.

Lead optimization in H2L relies on iterative structure-activity relationship studies to enhance desirable attributes while addressing potential liabilities. Scaffold modifications, bioisosteric replacements, and strategic incorporation of hydrogen bond donors/acceptors help fine-tune potency, selectivity, and PK properties. The application of ligand efficiency and lipophilic ligand efficiency metrics supports the selection of compounds that achieve potency gains without excessive increases in molecular weight or lipophilicity. Computational modeling and AI-driven predictive analytics further enhance the efficiency of H2L by providing insights into molecular behavior, protein-ligand interactions, and potential liabilities before extensive experimental validation.

A crucial consideration in H2L is early pharmacokinetic and pharmacodynamic evaluation, as in vitro potency alone does not guarantee in vivo efficacy. Pharmacokinetic profiling—including metabolic stability, plasma protein binding, permeability, and clearance—identifies compounds with favorable ADME properties. In parallel, early toxicological assessments help identify potential safety concerns before significant resources are invested in lead optimization.

The decision to progress or discontinue a chemical series is a key turning point in the H2L process. If a series fails to achieve an acceptable balance between potency, selectivity, and drug-like properties, it may be prudent to deprioritize it in favor of alternative scaffolds. Advanced analytics, including machine learning-guided predictive modeling, are increasingly utilized to streamline decision-making and reduce risk before committing to extensive medicinal chemistry efforts.

In conclusion, the Hit-to-Lead stage is a data-driven, multidisciplinary process that integrates medicinal chemistry, computational modeling, structural biology, and pharmacology to refine initial hits into viable lead compounds. Researchers can improve the likelihood of success in lead optimization and clinical development by optimizing key parameters such as potency, selectivity, pharmacokinetic properties, and synthetic feasibility.

To provide further insights, the authors have organized a webinar on H2L strategies, where experts will discuss best practices and real-world case studies. You can access the webinar at (https://www.efmc.info/hit-to-lead).

Open access article: https://doi.org/10.1002/cmdc.202400931

The European Federation for Medicinal Chemistry and Chemical Biology (EFMC) Best Practice Initiative: Hit to Lead, J. Quancard, A. Bach, C. Borsari, R. Craft, C. Gnamm, S. M. Guéret, I. V. Hartung, H. F. Koolman, S. Laufer, S. Lepri, J. Messinger, K. Ritter, G. Sbardella, A. Unzue Lopez, M. K. Willwacher, B. Cox, R. J. Young, ChemMedChem 2025, e202400931.

Sepideh Afshar, Ph.D.
Head of Peptide Therapeutics - Senior Director, Genentech
Member, BPF Scientific Advisory Board
linkedin.com/in/sepideh-afshar-bab3824

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 

Call for Nominations for the 2025 Young Investigator Award

The Boulder Peptide Foundation is now accepting nominations for the 2025 Waleed Danho Young Investigator Award. This award is given every two years to a young academic investigator with great potential for scientific contributions to the field of peptide chemistry, formulation, delivery, and analytical characterization.

The award recipient will speak at BPS 2025 in Boulder, CO on September 16, 2025.  The award winner will receive a $5000 USD honorarium in addition to paid travel, hotel, and registration, which will be covered by the Boulder Peptide Society.

The deadline to submit nominations is April 30, 2025.
To read about the award criteria visit https://www.boulderpeptide.org/young-investigator-award/

BPF Journal Club – March edition

AI Meets Peptides: Revolutionizing Therapeutic Discovery with Machine Learning

This month, we want to draw your attention to a review article by Goles et al. covering the field of artificial intelligence applied to peptide therapeutics (https://doi.org/10.1093/bib/bbae275).

This article provides a comprehensive exploration of the role of machine learning (ML) and artificial intelligence (AI) in advancing peptide drug discovery, with a strong focus on de novo design and optimization. The paper highlights several advanced ML techniques, including classifier methods for activity prediction, numerical property estimations, and deep generative models (DGMs) like generative adversarial networks (GANs) and variational autoencoders (VAEs) for sequence generation. Deep generative models are central to the de novo design of therapeutic peptides. VAEs, for instance, allow the encoding of peptide data into latent spaces to generate novel sequences resembling training data, while GANs iteratively refine the quality of generated sequences through adversarial training. Newer approaches, such as diffusion models, offer even higher fidelity in sequence generation by leveraging iterative, reversible transformations to fine-tune peptide designs. The use of predictive models alongside these generative techniques facilitates the assessment of physicochemical properties and binding affinities, streamlining the identification of promising candidates before experimental testing.

A unified AI-driven pipeline proposed in the article integrates several critical steps to optimize therapeutic peptide discovery. The process begins with functional classification models, which categorize peptide activity and evaluate properties. This is followed by affinity prediction models that assess peptide interactions with target proteins using deep learning architectures like graph convolutional networks. Generative methods then explore peptide sequence space, creating candidates with specific biological activities and favorable structural properties. The peptide properties are then optimized in silico for stability, toxicity, and pharmacokinetics. By
incorporating iterative validation and reinforcement learning, the pipeline allows continuous refinement of predictive and generative models using experimental feedback​.

Finally, the authors address the significant challenges that remain in advancing the field of predictive peptide design. One primary issue is the lack of centralized, high-quality peptide datasets, as fragmented and inconsistent data often lead to misclassification and hinder model training. Additionally, post-translational modifications (PTMs), critical to many of natural peptides' functional roles in vivo, are frequently overlooked in current design pipelines. Moonlighting effects, where peptides exhibit multiple biological activities, further complicate the prediction of specific properties. Addressing these challenges will enable the rapid and precise design of highly specialized peptides that can tackle a wide range of indications with unprecedented efficiency and specificity.

Open access article: https://doi.org/10.1093/bib/bbae275

Goles M., Daza A., Cabas-Mora G., Sarmiento-Varón L., Sepúlveda-Yañez J., Anvari-Kazemabad H., Davari M.D., Uribe-Paredes R., Olivera-Nappa A., Navarrete M.A., Medina-Ortiz D., Peptide-based drug discovery through artificial intelligence: towards an autonomous design
of therapeutic peptides, Briefings in Bioinformatics, Volume 25, Issue 4, July 2024.

Ewa Lis, Ph.D.
Founder/ CEO, Koliber Biosciences
Member, BPF Scientific Advisory Board
https://linkedin.com/in/ewa-lis-6b99528

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 

BPF Journal Club – December edition

Glucose-sensitive insulin – one step closer towards the “holy grail” of insulin research

With current insulin therapy there is still a risk of hypoglycemia, which in certain circumstances can be life-threatening. The quest for discovering a true glucose-sensitive insulin – an insulin, which can auto-adjust its bioactivity depending on actual glucose levels in vivo – has been a long-standing aspiration in pharmaceutical research. Sometimes it is even reflected as the “holy grail” of insulin research.

Several research strategies have been tried, including the release of insulin or insulin analogs from glucose responsive biomaterials as well as bioengineered insulins binding to glucose itself or glucose dependent proteins – so far with limited success and limited translatability from in vitro to in vivo systems. Very recently researchers from Novo Nordisk reported the discovery of a novel, smartly engineered insulin, NNC2215, which is reversibly glucose responsive to a glucose range relevant for diabetes, not only in vitro but also in vivo. “(https://www.nature.com/articles/s41586-024-08042-3)” This exciting new insulin was engineered by attaching a selective glucose binding macrocycle to the B29 Lys side chain and a glycoside to the B1 amino-terminus of DesB30 human insulin. These modifications enable the molecule to adopt distinct conformations depending on the glucose level – a bioactive “open” conformation at higher glucose levels (20 mM) and a less bioactive “closed” conformation at lower glucose levels (3 mM). Excitingly, NNC2215 could also demonstrate glucose responsiveness and protection from hypoglycemia when tested in diverse animal models (rats & pigs).

The approach reported here to engineer a truly reversible, glucose responsive insulin is one of the most promising achievements in the field and should be a “must read” for every peptide & diabetes researcher, from a drug discovery but also peptide design perspective.

Open access article: (https://www.nature.com/articles/s41586-024-08042-3)

Hoeg-Jensen T, Kruse T, Brand CL, Sturis J, Fledelius C, Nielsen PK, Nishimura E, Madsen AR, Lennart L, Halskov K, Koščová S, Kotek V, Davis AP, Tromans RA, Tomsett M, Peñuelas-Haro G, Leonard DJ, Orchard MG, Chapman A, Invernizzi G, Johansson E, Granata D, Hansen BF,
Pedersen TA, Kildegaard J, Pedersen KM, Refsgaard HHF, Alifrangis L, Fels JJ, Neutzsky-Wulff, Sauerberg P & Slaaby R. Glucose-sensitive insulin with attenuation of hypoglycemia Nature 634, 944 -951 (2024).

Michael Wagner
Dewpoint Therapeutics
Member, BPF Scientific Advisory Board
https://www.linkedin.com/in/michael-wagner-a086bb15/

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 

BPF Journal Club – November edition

A Sustainable Solid-Phase Peptide Synthesis Approach

This month, we want to draw your attention to the topic of sustainability in peptide synthesis. The article “In Situ Fmoc Removal – A Sustainable Solid-Phase Peptide Synthesis Approach” (Kumar et al., 2022), while a few years old, remains highly relevant. The authors pave the way for the concept of peptide sustainability and green chemistry by introducing a clever method to reduce solvent waste.

Until green solvents for solid-phase peptide synthesis (SPPS) become widely accepted in the industry, it's crucial to work towards minimizing the environmental impact of existing SPPS practices. This article addresses one of the largest sources of solvent consumption in solid peptide synthesis: the solvents used for resin washes.

The authors propose a method that combines acylation and Fmoc deprotection by directly adding neat piperidine to the spent solution containing residual amino acids and oxyma. This approach effectively telescopes the acylation reaction into the deprotection step, which they refer to as in situ Fmoc deprotection. This practical solution is exactly what the industry needs to tackle the short-term issues related to process mass intensity (PMI) in SPPS. The authors demonstrate that this method does not compromise the final product; they achieve peptide materials of equal purity compared to standard SPPS.

Another reason we selected this article is that its corresponding author and inspiration is Professor Fernando Albericio.

Professor Albericio has been a prominent figure in solid-phase peptide chemistry for the past 50 years, developing numerous reagents that are essential for peptide chemists in their daily syntheses. Recently, he has made significant contributions to green chemistry, focusing on environmentally friendly methods for peptide synthesis. Given the rising interest in peptides as potential drugs, such as GLP-1, his work is increasingly important. His dedication to replacing hazardous solvents like DMF, DCM, and NMP with safer, less toxic alternatives is consistently evident in his numerous recent publications on this topic.

Professor Albericio was recently honored with the Meienhofer Award for excellence in peptide sciences at the Boulder Peptide Symposium in Boulder, CO. His lecture was well received by attendees. His research emphasizes scaling up peptide synthesis for pharmaceutical applications, enabling the production of large quantities of peptides with minimal environmental impact. He has pioneered the use of greener solvents, such as water, achieving high yields and purity, and has conducted detailed studies on the properties of green solvents in SPPS. Additionally, he is exploring new protecting groups that are easily removed and generate less waste.

His career continues to inspire many students, and his commitment to green chemistry is a testament to his dedication to the field of peptide chemistry.

Kumar, A., Sharma, A., de la Torre, B.G., and Albericio, F. (2022). In situ Fmoc removal – a sustainable solid-phase peptide synthesis approach. Green Chemistry, 24, 4887-4896. Link to publication

Professor Pravin Kaumaya, Ph.D.
Indiana University School of Medicine
Member, BPF Scientific Advisory Board

Matteo Villain, PharmD.
Member, BPF Scientific Advisory Board
linkedin.com/in/matteo-villain

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 

Free Virtual BPS on Venom Peptides: Nov 13

The next Virtual BPF Seminar, "Discovering Therapeutic Applications of Snail Venom Peptides", will be held Wednesday, November 13th starting at 12:00 PM EST (9:00 AM PST). We will be hosting Dr. Mandë Holford (Professor, Hunter College - CUNY) and Dr. Walden Bjørn-Yoshimoto (Assistant Professor, University of Copenhagen). Each will present their work exploring therapeutic applications for snail venom peptides. A moderated Q&A session will follow each presentation.

Registration is free, and can be completed at Virtual BPS | Boulder Peptide Symposium. *Note* you must be signed in to the site in order to sign up.

These educational e-seminars are provided free of charge by the Boulder Peptide Foundation. If your company is interested in supporting this or other educational initiatives, please email sponsorship@boulderpeptide.org.

BPS 2024 Photos and Videos are available!

Thank you to everyone who attended and participated in the 2024 Boulder Peptide Symposium. We are pleased to announce that videos of the presentations from the Symposium are now available on our website for all of our attendees to view.  If you did not attend the meeting, you may purchase access by visiting the video subscription page. A gallery of photos from the event is also available on the website.  We look forward to seeing you at BPS 2025 back at the St. Julien Hotel in Boulder, CO beginning on September 15th, 2024.

CLICK HERE FOR VIDEOS

BPF Journal Club – October edition

Treating Fungal infections with synthetic peptide mimics

Modern medicine often relies on invasive medical interventions or drugs that can compromise the patient’s immune system. An unfortunate consequence of these undeniably successful treatments for life-threatening diseases like cancer is the occurrence of severe infections caused by opportunistic fungal pathogens, including species of Candida, Aspergillus, Cryptococcus, and Pneumocystis. More recently, increasing numbers of opportunistic fungal infections caused by Aspergillus, Mucorales, and Candida species have been observed in COVID-19 patients with severe respiratory syndromes in intensive care units. These factors, among others, have resulted in over 2 million invasive fungal infections annually worldwide, with alarmingly high mortality rates, causing more than 1.5 million deaths.

Candida species are the most common cause of nosocomial, invasive fungal infections and are associated with mortality rates above 40%. Despite the increasing incidence of drug-resistance, the development of novel antifungal formulations has been limited. In a recent publication by Schaefer, S., Vij, R., Sprague, J.L. et al., they highlight the enormous potential of synthetic peptide mimics to be used as novel antifungal formulations as well as adjunctive antifungal therapy.

Schaefer, S., Vij, R., Sprague, J.L. et al. A synthetic peptide mimic kills Candida albicans and synergistically prevents infection. Nat Commun 15, 6818 (2024). Link to publication

Trishul Shah
Global Director Sales and Marketing, PolyPeptide Group
Member, BPF Scientific Advisory Board
www.linkedin.com/in/trishul-shah-bb92a2/

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 

BPF Journal Club – September edition

Peptide Drug Conjugates: The New Frontier Beyond Antibody Horizons!

Targeted cancer therapy has seen a renaissance in recent years, particularly with radiopharmaceuticals.  Big Pharma has spent tens of billions in this niche field alone. But, peptides are ideally suited to be conjugated to many other types of warheads or generally active payloads, making the overall field of Peptide-Drug Conjugates (PDCs) exquisitely wide, and not limited to oncologic indications. These recent advances in PDCs seem to me (I admit, I have a preference for peptides) a logical step to overcome some of the complexity associated with ADC production and development. For example, peptides can be very well-controlled before and after payload conjugation with standard analytical techniques. Also, screening incredibly large and diverse peptide libraries for remarkable selectivity and specificity is a task that peptide researchers have mastered through a variety of innovative solutions. In this recent review on PDC recently published in the Journal of Medicinal Chemistry, the authors navigate the advantages of PDCs relative to their Antibody-Drug Conjugate (ADC) predecessors. They explore the various linkers, payloads, and overall design of PDCs, while presenting the next horizon for these modalities.  For any peptide chemist familiar with the recent tidal wave of macrocycle peptide discovery efforts, it should be clear there is a promising stage set to further enhance the appeal and potential for peptides in drug development.

Link to open access article: https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c00106

Zhang B, Wang M, Sun L, Liu J, Yin L, Xia M, Zhang L, Liu X, Cheng Y. Recent Advances in
Targeted Cancer Therapy: Are PDCs the Next Generation of ADCs? J Med Chem. 2024 Jul
25;67(14):11469-11487. doi: 10.1021/acs.jmedchem.4c00106

Christopher McGee, PhD
Member, BPF Scientific Advisory Board
www.linkedin.com/in/christopherjmcgee/

Read previous editions of the BPF Journal Club series: https://www.boulderpeptide.org/journal-club

 


s2Member®
loading...