Tag Archives: art

The Needle Issue #16

7 Oct
Juan-Carlos-Lopez
Juan Carlos Lopez
Andy-Marshall
Andy Marshall

Commercial interest in targeted epigenetic therapies — agents that target specific genes without altering bases in their sequence or causing double-strand breaks or even single nicks in the DNA — continues to grow, as underscored by the latest financing announced by Epigenic Therapies. The unique selectivity and specificity of targeted epigenetic therapeutics offers compelling advantages over small-molecule epigenetic drugs, which target a specific epigenetic reader, writer or eraser, but affect genes across the genome and affect many diverse tissues, leading to narrow therapeutic windows that make them difficult to develop for conditions outside of cancer.

Today, Haystack is aware of at least eight private companies (nChroma Bio (resulting from a merger of Chroma and Nvelop), Encoded TherapeuticsEpigenic TherapeuticsEpitor TherapeuticsMoonwalk BioNavega TherapeuticsRegel Therapeutics, and Tune Therapeutics), and two public companies (Modalis Therapeutics and Sangamo Therapeutics) that are pursuing the targeted epigenetic approach against disease (let us know if you know of any others). Another company, Flagship Pioneering’s Omega Therapeutics, went out of business in August after filing for bankruptcy in February. A smaller set of companies are also pursuing targeted epigenetic therapies against RNA modifications.

All of these therapies are designed around an alluring set of simple principles: take a gene-specific DNA-binding domain — zinc-finger proteins (ZFPs), ‘dead’ Cas9 (dCas9) with mutations in its RuvC and HNH endonuclease domains, or transcription activator-like effectors (TALEs) — and tether it via an amino acid linker to an enzymatic effector module. This effector is either an enzyme that directly places or removes a specific epigenetic modification (e.g., TEThistone demethylases or the histone acetyltransferase p300) or a transcriptional activator (e.g., VP16) or repressor (e.g., KRAB).

A particularly compelling application for such treatments is genetic disorders of haploinsufficiency (like Dravet’s) or imprinting disorders (like Angelman’s or Prader Willi). There are also many of these diseases where the therapeutic genes would be too large (>4.0 kb) for a traditional AAV gene-therapy approach; in contrast, epigenetic editing machinery can be packaged into an AAV vector.

Currently, the diseases being pursued by companies include hepatitis Bhypercholesterolemia, epilepsies (SCN1A (Dravet syndrome) and SCN2A), chronic pain, and muscular dystrophies. Those with the most advanced programs are Encoded’s AAV-9 intrathecally delivered SCN1A-targeting zinc finger protein linked to a VP16 activation domain in phase 1 testing for Dravet and Sangamo’s AAV- STAC-BBB-delivered SCN9A-targeting zinc finger protein linked to a KRAB repressor domainin a phase 1/2 trial for patients with chronic pain. In this context, two papers published in the past couple of weeks represent important proofs of the efficacy of targeted epigenetic therapies.

In a first paper published in Nature, the groups of Kevin Bender and Nadav Ahituv at UCSF (scientific co-founders of Regel Therapeutics) sought to test a targeted epigenetic therapy in patients with SCN2A mutations that exhibit decreased NaV1.2 function. These individuals have impaired action potentials, synaptic transmission and manifest diverse neurological symptoms and seizures, with few therapeutic options, beyond symptomatic anti-seizure medications that have a dizzying range of debilitating side effects.

The UCSF teams leveraged conditional genetic knock-in technolgoy or CRISPRa technology — an AAV-delivered SCN2A-promoter-targeting dCas9 fused to a VP16 activator domain — to upregulate transcription of the SCN2A gene. Using either approach, they were able to boost transcript levels from the healthy SCN2A allele, ameliorating electrophsiological deficits and chemical-induced seizure activity in Scn2a+/− mouse models. Importantly, these effects were seen in adolescent mice, which conventionally have been thought to be too old to respond to treatment. This suggests that rescue of normal dendritic excitability with epigenetic agents at later stages of life might be capable of restoring neuronal function, with implications for patients.

In a separate set of experiments, the authors showed that their epigenetic approach was able to rescue neurophysiological activity in haploinsufficient neuron-like cells from SCN2A-knockout human embryonic stem cells. This cross-species reproducibility provides further confidence that CRISPRa-mediated upregulation could be translated into human treatments.

In a second paper in Nature Biotechnology, a team from Epigenic Therapeutics (Shanghai, China) describes the design and validation of optimized epigenetic regulators (EpiRegs) to silence genes in a precise, durable way without altering genomic DNA. Epigen’s Shaoshai Mao and his collaborators at the Chinese Academy of Sciences and the First Affiliated Hospital of Anhui Medical University tested combinations of TALE- and dCas9-based systems, systematically optimizing effector domains and fusion architectures, looking for effective regulators of gene expression. The best-performing variant, EpiReg-T (a TALE-based system, which eliminates the need for a guide RNA), achieved 98% silencing of target genes in mice, substantially outperforming dCas9-based versions.

Using lipid nanoparticles (LNPs) for delivery, a single administration of EpiReg-T in macaques induced long-term repression of the PCSK9 gene, which encodes a validated target for the treatment of hypercholesterolemia. EpiReg-T reduced PCSK9 expression by >90% and LDL-cholesterol by about 60%, with effects persisting for nearly a year (343 days).

Mechanistically, the team used whole-genome bisulfite sequencing and cleavage under targets and tagmentation (CUT&Tag) to show that EpiReg-T induced stable DNA methylation and repressive histone marks at the PCSK9 promoter. The silencing persisted even after liver regeneration and could be reversed by targeted epigenetic activation. Multiomic analysis in mice, macaques and human hepatocytes confirmed high specificity of the manipulation and minimal off-target effects. Overall, these finding, as well as similar results reported in April by Chroma Medicine, establish epigenetic editing as a promising therapeutic platform for durable and reversible gene silencing.

Overall, targeted epigenetic therapies offer clear safety advantages over small molecules that indiscriminately target all genes under the control of an epigenetic eraser or writer enzymes. They avoid the potential risks associated with creating single- or double-strand DNA breaks associated with CRISPR/Cas9 gene, base or prime editing therapies. And they avoid the insertional mutagenesis risks associated with traditional viral gene therapies. What’s more, in applications requiring gene upregulation in haploinsufficient disease, these approaches maintain the endogenous regulatory context of the functional allele. This is in stark contrast to traditional gene-therapy replacement approaches, where overexpression of an introduced therapeutic gene can often lead to toxicities and immunogenecity.

Of course, questions still linger around the persistence of the changes elicited by these epigenetic agents. Will they persist in patients for long periods — for years or even decades? If they can, then epigenetic therapy may offer compliance advantages over small molecules, antibodies, ASOs or even siRNAs, which have treatment durations of six months or less.

Like all genetic medicines, though, delivery remains the key headache. Thus far, AAV vectors, lipid nanoparticles or ribonucleoproteins (RNP) have all been explored to deliver epigenetic therapies (with some evidence that RNPs might have advantages because they can result in higher dCas9 dosages within target cells). For AAV vectors, the fact that targeted epigenetic therapy might only need to be given once might be an advantage in terms of immunogenicity/neutralization concerns against the vector.

A broader point is that the safety profile of targeted epigenetic editors may offer advantages if AAV vectors are used as delivery vehicles: if the epigenetic agents themselves can be delivered at high dosage (given their intrinsic favorable safety profile and presumed maximal tolerated dose), perhaps AAV vector dosages could be lower than current practice. With many current gene therapies requiring dosages of 1013 or more viral particles/kg in patients, it is increasingly becoming clear that unacceptable liver toxicities arise from the virus at these levels in clinical studies. It will be interesting to follow this space as more agents enter human testing.

Super Early Bird Rates End October 3 for RESI London and RESI JPM 

30 Sep

By Sougato Das, President and COO, LSN

Sougato-Das

Life Science Nation’s Redefining Every Stage of Investments (RESI) conferences are designed to connect life science companies with global investors and strategic partners. Two major upcoming events, RESI London and RESI JPM, are now open for registration at Super Early Bird rates through October 3. By registering early, attendees can save £300 on RESI London and $800 on RESI JPM.

RESI London 2025

The first week of December marks the largest life science partnering and venture week in the UK. If you are raising pre-seed, seed, round A or round B, or are in phase 2 or earlier development, Life Science Nation’s RESI London, One Nucleus’ Genesis, and ELRIG/SLAS events offer stronger partnering, investment, learning, and procurement opportunities compared to the health care week in late November that runs alongside the Jefferies investment banking event. That November week typically focuses on recent IPOs, companies close to IPO, and phase 3 or more advanced companies seeking partnerships. For companies not yet at that stage, December’s conference series is a more strategic use of critical conference and travel budgets.

RESI London and Genesis are joining to provide a multi-day investment, partnering, and thought leadership event for venture-stage companies pursuing funding and strategic alliances. RESI London will take place on December 4 at 1 Wimpole Street and 11 Cavendish Square, followed by two days of virtual partnering on December 5 and 6. Super Early Bird registration is now open, offering a savings of £300 until October 3.

RESI JPM 2026

RESI JPM will be held January 12–13, 2026, at the San Francisco Marriott Marquis. The program features two days of in-person investor panels, workshops, networking, and one-on-one partnering, followed by three days of online partnering on January 14, 19, and 20.

Previous RESI JPM conferences have drawn more than one thousand attendees, including over 500 investors, innovators, and industry experts from across the globe. The conference is held concurrently with JP Morgan Healthcare Week, January 12–15, 2026, which takes place at the Westin St. Francis. Known as the largest healthcare investment symposium worldwide, JPM attracts thousands of life science professionals each year.

Super Early Bird registration for RESI JPM offers a savings of $800 when completed before October 3.

Register for RESI London
Register for RESI & Genesis Combi Ticket
Register for RESI JPM

Both conferences provide access to global investors and in-licensors, strategic partners, and hundreds of early-stage innovators across therapeutics, medical devices, diagnostics, and digital health. Register now to take advantage of Super Early Bird discounts for RESI London and RESI JPM before October 3.

LSN Staff on Global Event Circuit; Find Out Who, What, and Where

30 Sep

By Max Braht, Director of Business Development, LSN

Max-Braht-Headshot

Life Science Nation is on the move. Between now and RESI London, our team will be attending a series of leading life science and medtech conferences around the world, connecting with innovators, investors, and strategic partners. Getting out into the global ecosystem allows us to better understand emerging trends, foster collaborations, and support life science companies in reaching their fundraising and partnership goals.

These engagements also reinforce the value of our global conference, RESI, which brings together investors, entrepreneurs, and strategic partners from across the world to drive innovation in life sciences. Attending regional and international events helps us strengthen these connections and grow our global community.

Here’s where you can find our team in the coming months:

October 
Oct. 4–8: San Diego for the MedTech conference, engaging with medical device innovators and investors. San Diego, CA
  • Max Braht (m.braht@lifesciencenation.com)
  • Matt Stanton (m.stanton@lifesciencenation.com)
Oct. 4–8: LSN Bootcamp and BioSpain, connecting with early-stage life science companies. Barcelona, Spain
  • Greg Mannix (g.mannix@lifesciencenation.com)
  • Karen Deyo (k.deyo@lifesciencenation.com)
  • Brenda Olmos (b.olmos@lifesciencenation.com)
Oct. 8–10: Japan for BioJapan, focusing on international biotech partnerships. Yokohama, Japan
  • Dennis Ford (dford@lifesciencenation.com)
  • Claire Jeong (c.jeong@lifesciencenation.com)
  • Momo Yamamoto (m.yamamoto@lifesciencenation.com)
Oct. 21-23: Sao Paulo for Corporate Venture in Brazil 2025, highlighting innovation and investment opportunities in Latin America. Sao Paulo, Brazil
  • Dennis Ford (dford@lifesciencenation.com)
  • Max Braht (m.braht@lifesciencenation.com)
  • Matt Stanton (m.stanton@lifesciencenation.com)
Oct 24-25: Washington DC for the 9th Annual KAPAL Conference, meeting global leaders in biotech and healthcare. Washington DC
  • Sougato Das (s.das@lifesciencenation.com)
Oct. 27–28: Miami for the BioHunt Summit, meeting emerging life science startups and investors. Miami, FL
  • Dennis Ford (dford@lifesciencenation.com)
  • Matt Stanton (m.stanton@lifesciencenation.com)
November 
Nov. 2–6: Vienna for BioEurope, connecting with corporate and venture partners across the life science ecosystem. Vienna, Austria
  • Max Braht (m.braht@lifesciencenation.com)
  • Greg Mannix (g.mannix@lifesciencenation.com)
Nov. 12–14: Malta at the MedTech World Summit, exploring opportunities in medical technology. Valletta, Malta
  • Greg Mannix (g.mannix@lifesciencenation.com)
  • Brenda Olmos (b.olmos@lifesciencenation.com)
Nov. 16–20: London for Jefferies London Life Sciences Week, engaging with global investors and biotech innovators. London, UK
  • Max Braht (m.braht@lifesciencenation.com)
  • Greg Mannix (g.mannix@lifesciencenation.com)
December 
Dec. 2: France for Biofit/Medfit, meeting early-stage innovators and strategic partners in biotech and medtech. Strasbourg, France
  • Max Braht (m.braht@lifesciencenation.com)
  • Matt Stanton (m.stanton@lifesciencenation.com)

We look forward to seeing our community at these events. If you’d like to connect with us while we’re on the road, please reach out to resi@lifesciencenation.com

Finalist in the RESI Boston Innovator’s Pitch Challenge – Meet M6P Therapeutics

30 Sep

In this interview, Caitlin Dolegowski speaks with Cuong Do, Founder and Chairman of M6P Therapeutics, about the company’s groundbreaking lysosomal targeting platform, its applications in rare disease and oncology, and the experience of pitching at RESI Boston.

Cuong Do
CaitiCaitlin Dolegowski

Caitlin Dolegowski (CD): M6P Therapeutics has achieved what was long thought impossible, delivering proteins to lysosomes. Can you explain the significance of this breakthrough?

Cuong Do (DO): An enzyme called GlcNac-1-phosphotransferase (PTase) is responsible for adding mannose 6-phosphate to the surface of lysosomal enzymes. People have tried and failed for decades to increase the expression of M6P, and everybody gave up. Our co-founder Stuart Kornfeld never gave up. He and his post-doc were able to engineer a variant of PTase that turned out to be 20X more effective than PTase itself in adding M6P to lysosomal enzymes. We built upon this breakthrough to create a platform that is able to create enzyme replacement therapies that have very high M6P content. Furthermore, our gene therapies are the only ones that result in M6P-containing enzymes being produced by the transduced cells.

We expanded upon the innovation and created chimeric antibodies that contain M6P as well. This allows these antibodies (after they bind to the targeted antigens) to be brought to lysosomes in virtually all cells in our bodies for degradation. This is a significant advantage over traditional antibodies relying on Fc clearance by only select immune cells.

CD: You have multiple rare pediatric drug designations and two programs nearing the clinic. What are the most exciting upcoming milestones for your pipeline?

DO: We are preparing to start an Investigator Initiated Trial in Australia for our M021 ERT for Pompe Disease in hopes of obtaining early human data demonstrating M021’s superiority over the standard of care.

CD: How does your lysosomal targeting platform extend beyond rare diseases, particularly in oncology with your chimeric PD-L1 and PD-1 antibodies?

DO: We figured out a way to add M6P to any protein, including antibodies. Our chimeric antibodies can be cleared by virtually all cells in the body since virtually all cells have receptors for M6P. This is especially effective for clearing surface antigens from cell surfaces. Our chimeric PD-L1 antibody is able to clear virtually all PD-L1 from the surface of tumor cells and thus activate T-cells and drive T-cell mediated tumor killing. Our chimeric version of Keytruda is able to remove PD-1 from the surface of T-cells and has shown to be more effective in inhibiting tumor growth in vivo than Keytruda itself.

CD: Can you walk us through your IP position and how it supports your growth strategy?

DO: We have invested heavily in IP that has created a portfolio of 9 patent families, 9 issued patents, and ~20 still in prosecution.

CD: Where are you in your fundraising journey, and what types of investors or partners are you looking to engage with?

DO: We have raised ~$40 million in our Seed and A rounds, which we invested to get our programs to where they are today. We are trying to raise a $5 million bridge now in anticipation of a $50+ million Series B next year. In addition to investors, we want to engage with potential partners who might be interested in our molecules.

CD: How did participating in the Innovator’s Pitch Challenge at RESI Boston help advance your business development or investor connections?

DO: We met a few companies who might be interested in partnering on some of our molecules. We’re continuing the conversations.


IPC Applications are now open for the next Innovator’s Pitch Challenge at RESI London 2025 and RESI JPM 2026, with spots filled on a rolling basis.

The Needle Issue #15

23 Sep
Juan-Carlos-Lopez
Juan Carlos Lopez
Andy-Marshall
Andy Marshall

On September 11, the Lasker Foundation awarded the 2025 Lasker~DeBakey Clinical Medical Research Award to Michael Welsh, Jesús González and Paul Negulescu for discoveries that led to the development of Trikafta, a triple combination of cystic fibrosis transmembrane conductance regulator (CFTR) potentiators and correctors to treat cystic fibrosis. This award recognizes the contribution of Trikafta to improving the quality of life of ~90% of the 40,000 people living with this condition in the United States, reducing infection-related hospitalizations and lung transplants, among other benefits.

But what about the other 10% of patients who don’t respond to Trikafta, many of whom carry so-called Class I alleles that cannot be rescued by this drug combination? Although a lot of progress has been made, several obstacles lie in the path of effective medicines for people who produce no, or negligible amounts of, CFTR protein.

It should come as no surprise that the main therapeutic strategies for Class I alleles aim to put missing CFTR back into lung cells. Among these strategies, mRNA delivery is the most advanced. VX-522, an RNA therapeutic program from Vertex and Moderna currently in Phase 2, is an inhaled drug that aims to deliver full-length CFTR mRNA to the lung using lipid nanoparticles (LNPs). Two related, competing mRNA delivery programs are at a similar stage of clinical development: ARCT-032 by Arcturus Therapeutics using their LUNAR LNPs; and RCT-2100 by ReCode Therapeutics, which uses a lung-targeted SORT (selective organ-targeting) LNP.

A key feature of RNA-based therapies is that any therapeutic benefit would likely be transient, requiring periodic administration of the medicine to achieve sustained effects. Gene therapy and gene editing have the potential to be a curative, “one and done” procedure. Thus far, however, only gene therapy programs have advanced far enough to be in human testing.

Of these, 4D Molecular Therapeutics’ 4D-710 and Spirovants’ SP-101 use different AAV subtypes designed to optimize delivery to airway basal epithelial cells of a CFTR minigene that lacks the regulatory domain. Both projects are in Phase 1/2 of clinical development.

As the large size (6.2 kb) of the CFTR transgene exceeds the packaging capacity of AAV vectors, Krystal Biotech and Boehringer Ingelheim have launched Phase 1/2 clinical programs using viral vectors with a greater payload capacity: KB407 is a re-dosable herpes simplex virus (HSV)-1 vector with a cargo capacity >30 kb that delivers two copies of the CFTR gene to lung epithelial cells using a nebulizer. BI 3720931 is Boehringer’s inhaled lentiviral vector pseudotyped with Sendai virus F and HN envelope proteins (rSIV.F/HN) engineered to deliver a single copy of the CFTR gene. Further behind in the pipeline, Carbon Biosciences’ CGT-001 is a nebulized non-AAV parvovirus-based vector capable of delivering full-length CFTR gene. Thus far, it has been tested in nonhuman primates and in human bronchial cells in culture.

Companies are also pursuing oligonucleotide therapies to modify disease-causing mutations at the RNA level. SPL84 is an inhaled antisense oligonucleotide (ASO) addressing a splicing defect (cryptic exon; class V mutation) in the ~1,600 CF patients who carry the 3849+10kb C→T mutation. SpliSense has advanced the ASO into phase 2 testing, but it also has in preclinical development an exon-skipping ASO against the class I mutant W1282X. By masking the mutant premature termination codon in exon 23, SP23 induces the splicing machinery to skip exon 23 and stitch together exon 22 and exon 24, forming a partially functional CFTRΔex23 protein.

Gene editing is also beginning to appear on the therapeutic horizon. In July, Prime Medicine announced it had received $25 million in funding to advance prime editors, with a lead program focusing on G542X. Last year, Intellia Therapeutics and ReCode Therapeutics also announced a strategic collaboration to combine the CRISPR pioneer’s Cas9 DNA ‘writing’/insertion technology with Recode’s SORT LNPs. Academic groups have now shown that G542X correction is possible using inhaled LNP- or virus–like particle-delivered adenine base editors. And for RNA editing, at this year’s American Society of Gene & Cell Therapy Wave Life Sciences reported their oligo-based ADAR editors could achieve 21% correction (EC50 = 376nM) of CFTR W1282X nonsense mutations. This is likely a sliver of all the therapeutic activity underway; other programs are targeting mucus itself, which is much thicker than in healthy individuals. If we missed any drug-discovery projects in this space, please let us know!

Despite the plethora of programs, developing genetic therapies against cystic fibrosis patients with class I CFTR mutations faces some stiff translational challenges. For starters, targeted delivery of drugs to lung tissue remains a work in progress. The optimal cell type to be targeted by gene therapy/editing remains an open question, especially as the community continues to identify new cell types in the lung; is it enough to target the more prevalent epithelial cells (alveolar type 2 cells), or will it be necessary to target rarer stem cells (alveolar type 1 cells) to see a long-lasting therapeutic effect? What about the contribution of genetic modifiers and other ion channels known to affect airway dysfunction in CF airway epithelial cells? Also, how to figure out the pharmacokinetics and pharmacodynamics of these disease-modifying therapies in lungs and measure delivery in patients? Specifically, establishing protein expression levels after inhaling a DNA- or RNA-based product would likely require a bronchial biopsy, which is impractical particularly in this fragile patient population.

Last, not unlike most pathologies, new animal and in vitro models with predictive value need to be developed. The use of human bronchial epithelium culture is not as predictive of the efficacy of genetic therapies as it has been for small molecules. At present, the ferret is the gold standard disease model. But it is a time-consuming, challenging animal model, which is only supported by a few groups. All of which slows the path to clinical translation.

Six years after the approval of Trikafta, patient foundations like the CF Foundation, Emily’s Entourage, and the Cystic Fibrosis Trust are devoting increasing resources to translational research to push forward treatments for patients with CFTR Class I mutations who do not respond to potentiators and correctors. The Lasker recognition of the science that led to Trikafta will surely inspire researchers working on those projects to overcome the remaining hurdles.

Partnering for Growth: DLA Piper on Supporting Life Science Innovation at RESI Boston 

9 Sep

At RESI Boston, global law firm DLA Piper plays a key role in guiding early-stage innovators through the legal and commercial challenges of scaling in the life sciences. In this interview, Lauren Murdza, Co-Chair of Technology & Life Sciences Licensing & Commercial Transactions, shares why DLA Piper chose to sponsor RESI, what the firm looks for in collaborations, and the trends shaping licensing and commercial transactions today.

Lauren Murdza
CaitiCaitlin Dolegowski

Caitlin Dolegowski (CD): What motivated DLA Piper to sponsor RESI Boston, and why do you see value in supporting this conference?

Lauren Murdza (LM): DLA Piper is committed to supporting innovation in the life sciences sector, and RESI Boston offers a unique opportunity to engage directly with early-stage companies and investors. Sponsoring RESI aligns with our mission to be a strategic partner to emerging life science ventures, helping them navigate legal complexities while fostering meaningful connections that drive growth.

CD: From your perspective, what makes RESI a strong platform for connecting with early-stage life science innovators and investors?

LM: RESI creates a unique environment where entrepreneurs, investors, and advisors come together to solve real challenges. For DLA Piper, it’s an opportunity to listen and engage in conversations that matter—how to protect IP, manage data rights, and structure collaborations that attract capital. Those discussions allow us to show how DLA Piper’s integrated approach—combining legal, regulatory, and commercial insight—helps companies accelerate their next milestone.

CD: Can you share what types of companies, technologies, or partners DLA Piper is most interested in engaging with during RESI?

LM: We’re particularly interested in companies developing novel therapeutics, diagnostics, digital health platforms, and medical devices. Our team seeks to engage with founders and executives who are navigating the transition from concept to commercialization and who value strategic legal guidance in areas such as licensing, IP protection, and regulatory compliance.

CD: How does your team at DLA Piper support life science and healthcare companies as they move from early-stage development to commercialization?

LM: DLA Piper supports clients across the full lifecycle of a company—from corporate formation and IP strategy to licensing, financing, and M&A. We help clients identify the core aspects of their technology, assess patentability, and streamline initial filings to create contingent assets that support fundraising. What sets DLA Piper apart is our ability to deliver this seamlessly across jurisdictions, giving clients the confidence that their legal strategy scales with their business.

CD: Are there particular trends or challenges in licensing and commercial transactions that you think entrepreneurs at RESI should be especially mindful of?

LM: We’re seeing three big themes. First, clarity on data and AI rights is critical—investors want to know who owns what and how data can be used, especially across borders. Second, deal structures are evolving, with more options-to-license, milestone-based terms, and royalty monetization to help bridge funding gaps. Finally, regulatory and supply chain issues—from FDA expectations to manufacturing scale-up—are showing up earlier in negotiations. At DLA Piper, we help clients anticipate these challenges so they don’t slow down growth.

CD: What does DLA Piper hope to accomplish through its participation at RESI Boston this year?

LM: We aim to deepen our engagement with the life sciences community, share actionable insights through workshops and panels, and identify promising companies that could benefit from our legal and strategic expertise. RESI Boston is a chance to listen, learn, and contribute to the ecosystem that’s shaping the future of healthcare innovation.

CD: Looking ahead, what excites you most about the current life science innovation landscape, and how does DLA Piper plan to play a role in advancing it?

LM: We’re excited by the convergence of AI, data science, and biotechnology, which is accelerating discovery and personalization in medicine. DLA Piper plans to continue supporting innovators by offering forward-thinking legal solutions and fostering connections that help companies bring transformative technologies to the market.

The Needle Issue #14

9 Sep
Juan-Carlos-Lopez
Juan Carlos Lopez
Andy-Marshall
Andy Marshall

It is now nearly a decade since Dublin-based startup Inflazome burst onto the scene with the description of MCC950, the first nanomolar selective inhibitor of the inflammasome. Inflammasome-mediated low-grade inflammation has been associated with cancers, numerous chronic complex diseases—including inflammatory bowel diseasearthritismetabolic dysfunction-associated steatohepatitis (MASH)atherosclerosisAlzheimer’sParkinson’s and ALS—as well as rare autoinflammatory diseases, such as cryopyrin-associated periodic syndromes (CAPS). There is a wealth of compelling human genetic evidence from Muckle-Wells syndrome and other autosomal dominant familial genetic diseases showing gain-of-function mutations in NLRP3 are causative of autoinflammatory disease.

Given the ‘pipeline in a product’ potential of drugs targeting this pathway, big pharma has shown considerable interest, with Genentech/Roche snapping up Jecure Therapeutics for an undisclosed amount, and both Novartis and Roche splashing out hundreds of millions of dollars for pioneer companies IFM Tre and Inflazome, respectively. In 2022, Novo Nordisk licensed Ventus Therapeutics’ peripherally restricted NLRP3 inhibitor in a deal worth up to $703 million, lending weight to pharmacological inhibition of NLRP3 as a complement to glucagon-like peptide-1 agonists (GLP-1s) in cardiometabolic disease. And with several programs now entering the clinic, investment activity in the area has continued, with Enveda’s announcement last week of a $150 million series D round to fund a phase 1 trial for ENV-6946, an orally delivered gut-restricted small molecule targeting the NLRP3/tumor necrosis factor-like cytokine 1A (TL1A) pathway in inflammatory bowel disease.

Today, Haystack counts at least 17 independent companies pursuing inflammasome therapeutics (AC ImmuneAzome TherapeuticsBioAge LabsCardiol TherapeuticsEpicentRxEnvedaHalia TherapeuticsInflammX TherapeuticsInsilico MedicineNeumora TherapeuticsNodTheraOlatec TherapeuticsShaperonVentus TherapeuticsVentyx BiosciencesZyVersa Therapeutics and Zydus LifeSciences) and 8 programs now in clinical testing specifically targeting the key inflammasome component NLRP3.

While drugmakers have traditionally targeted downstream extracellular mediators of the inflammasome pathway (canakinumab or rilonacept against IL-1β or anakinra to block IL-1 receptor), NLRP3 represents a key upstream intracellular signaling hub, activated by innate immune pattern-recognition receptor (Toll like receptors 2/4) signaling via MyD88 and NFkappaB. Once activated, NLRP3 monomers unfold and associate into a massive 1.2 MDa oligomeric supracomplex with three other proteins: ASC, NEK7 and caspase 1. The mature complex then cleaves and activates proinflammatory cytokines interleukin (IL)-1β and IL-18 and primes gasdermin D to instigate cell pore formation and cell death via pyroptosis.

Discovering effective drugs against NLRP3 has proven challenging. The first NMR structure was obtained in 2016, but the structural basis for how NLRP3 ring-like oligomers associate with intracellular membranes and how its pyrin domains associate with ASC to orchestrate speck formation and caspase activation have only recently been elucidated. Thus far, the majority of small-molecule inhibitors (e.g., Inflazyme’s archetypal MCC950 and inzolemidZydus’s (ZYIL1)Olatec’s OLT117 and Jecure Therapeutics’ GDC-2394) form hydrogen bonds via a sulfonylurea group to NLRP3’s NACHT domain nucleotide-binding motifs, thereby obstructing ATP hydrolysis. Other companies are taking a different tack: thus, Halia Therapeutic’s small-molecule inhibitor orniflast and Monte Rosa Therapeutics’ MRT-8102 molecular glue target NEK7 rather than NLRP3.

But it has been less than straightforward to identify compounds with sufficient potency to target this pivotal innate immune signaling pathway without debilitating off-target effects. Indeed, several of the first wave of compounds entering the clinic have been dogged by serious toxicities, including liver problems (MCC950 and GDC-2394) and hypoglycemia (glyburide). Now, a team led by Rebecca Coll (Queen’s University Belfast) and Kevin Wilhelmsen (of BioAge Labs) reports in The Journal of Experimental Medicine the discovery and characterization of BAL-0028, a novel and selective small-molecule inhibitor of the human NLRP3 inflammasome.

Unlike previously studied inhibitors, BAL-0028 acts through a unique mechanism of action; it binds NLRP3’s NACHT domain at a site distinct from other inhibitors that act by directly interfering with ATPase activity. BAL-0028 has nanomolar potency against human and primate NLRP3 but, remarkably, has weak activity against the mouse target, highlighting species-specific differences.

As BAL-0028 showed very high plasma protein binding in mice, limiting its use in vivo, the team developed a derivative, BAL-0598, with improved pharmacokinetic properties. In a humanized NLRP3 mouse peritonitis model, BAL-0598 effectively reduced IL-1β and IL-6 production, confirming its anti-inflammatory activity in vivo. Importantly, both BAL-0028 and BAL-0598 inhibited hyperactive NLRP3 mutants associated with autoinflammatory diseases, in some cases more effectively than Vertex’s VX-765, a caspase 1 inhibitor, and compounds like MCC950, one of the best characterized NLRP3 inhibitors available.

The novel mechanism of action of BAL-0028 and BAL-0598 would suggest their off-target effects may be different from those associated with other NLRP3 inhibitors blocking ATP hydrolysis. The concern that such compounds might also bind other members of the NOD/NLR family (e.g., NLRP1, NLRP4 or AIM2 inflammasomes) is mitigated by most published studies indicating that NLRP3’s unique fold around the ATP binding site makes small-molecule binders selective for this family member alone. The most likely explanation from trials published to date is that the observed toxicities are associated with small molecule chemotype rather than any NLRP3 class-specific problem. In any case, the findings from this study support further investigation of these compounds as candidates for treating inflammatory and age-related diseases where NLRP3 plays a role. The race to develop a safe and effective NLRP3 inhibitor is on, with big pharma billion-dollar bets and startups jostling to create best-in-class assets across cancer, cardiovascular, neurodegenerative and metabolic disease.