The Needle Issue #7


Welcome to the latest issue of the Needle, a newsletter from Haystack Science on the latest translational research and goings on in the biotech startup world.

This week, we take a look at an exciting breakthrough in hematopoietic stem cell (HSC) gene therapy using lentiviral vectors injected directly into the body. There was an unusually large slew of interesting therapeutic papers, including a description from Insilico Medicine of the discovery of an asset from its internal AI-potentiated drug discovery program that has made it to clinical testing. We also take a look at startups presenting data at the American Society for Clinical Oncology and roundup the rest of the week’s news, including a new tranche of funding for cancer startups. The deal front for preclinical startups was very quiet this week.

Haystack chat

Ex vivo HSC lentiviral gene therapies have been on the market for nearly a decade, with six products approved and at least 55 now in clinical testing for rare inherited diseases, HIV infection or cancer. And yet, their commercial success remains in question. Bluebird Bio—which was valued at $10 billion only a few years ago and successfully shepherded to market Zynteglo against transfusion-dependent β-thalassemia, Skysona for early cerebral adrenoleukodystrophy, and Lyfgenia for sickle-cell disease (SCD)—was sold earlier this year to private-equity firms Carlyle and SK Capital for a measly $29 million. Last November, the company had treated only 57 patients (35 for Zynteglo; 17 for Lyfgenia and 5 for Skysona), with just 28 of 70 medical centers across the US ready to treat patients due to delays in accreditation and training of personnel. In Europe, Orchard Therapeutics halted marketing and production of a treatment for severe combined immunodeficiency caused by adenosine deaminase mutations (Strimvelis) after six years, forcing Fondazione Telethon to take over production. Even market uptake of Vertex’s much-heralded CRISPR/Cas9 BCL11a SCD therapy Casgevy has been sluggish.

These subpar commercial launches relate to the complexity of ex vivo lentiviral gene therapy: patient identification and qualification is lengthy; HSC mobilization and sourcing efficiencies vary due to patient heterogeneity; and manufacture and distribution processes remain lengthy and convoluted (sometimes requiring repetition if a poor quality product batch is generated). From first evaluation, patients are required to make several hospital visits over a period (of up to a year) and must undergo punishing conditioning regimes with lymphodepletive bisulfan before infusion, which itself carries infertility and cancer risks. All of these challenges have added impetus to the search for alternative and more efficient approaches for carrying out HSC gene therapy.

A group led by Alessio Cantore and Luigi Naldini, from the San Raffaele Telethon Institute for Gene Therapy in Milan, Italy, report in Nature that it may be possible to obviate these challenges by delivering recombinant lentiviral vectors in vivo soon after birth, when HSCs continue to circulate in the bloodstream in large numbers and are beginning their transition from the liver (where they are located in the fetus) to bone marrow (where they remain through adulthood).

Cantore, Naldini and their colleagues started by measuring the number of circulating HSCs in neonatal, 1-, 2- and 8-week-old mice, looking at the peripheral blood, spleen, liver and bone marrow. They found that HSCs were present in the circulation right after birth and that their number immediately declined. These cells could be transduced with lentiviruses, successfully engrafted, and persisted in the mice for several months.

To show that these HSCs could be harnessed to treat genetic disorders, the team tried to correct three mouse models of disease — adenosine deaminase deficiency, autosomal recessive osteopetrosis and Fanconi anemia. Although the therapeutic effect of the cells varied depending on the disease, the results provided compelling evidence for the potential for in vivo gene transfer to HSCs.

The authors reported that human neonates also have circulating HSCs in high numbers. And although the therapeutic window in the mouse only existed during the neonatal period, it was possible to lengthen it by mobilizing the HSCs from their niche in two-week-old animals using protocols in clinical use (granulocyte-colony stimulating factor/CXCR4 antagonist Plerixafor) These observations raise the possibility of therapeutically targeting HSCs in newborns, potentially opening the gates to treatment of a variety of inherited conditions.

Compared with the headaches of ex vivo manipulation, the authors’ concept of simply injecting a lentiviral gene therapy into a newborn to bring about a genetic cure is certainly alluring. But reducing this to clinical practice will require optimization of many different factors. How to account for the heterogeneity and fragility of patient HSCs in a particular disease? How to measure the cellular activation/metabolic state of HSCs in newborns and assess the affect on amenability to lentiviral transduction in the hostile milieu of blood? What effect would shear stress in circulation have on lentiviral transduction efficiencies in situ? What would be the selective engraftment advantage provided to HSCs after engraftment of a particular gene? And what would be the potential safety implications of off-target transduction events in cells other than HSCs, given instances of dysplastic syndromes have been reported with ex vivo lentivectors?

Current ex vivo lentiviral gene therapy like Lyfgenia and Zynteglo infuse between 3–5×106 gene-modified CD34+ HSCs/kg in a patient. The challenge for in vivo lentiviral gene therapy will be to achieve transduction efficiencies that transduce as many cells and obtain similar engraftment rates in the rapidly turning over HSC population. Beyond these issues, there are additional practical challenges: can genetic testing of an infant happen fast enough to take advantage of the short therapeutic window for which an in vivo lentiviral HSC therapy could work?

Clearly, the new work raises many intriguing questions for the lentiviral gene therapy space. And for newborns with genetic diseases, such as severe immunodeficiencies or Fanconi anemia, in vivo HSC gene therapy may open up new treatment options.

Papers: Best of the rest

Target biology

Skeletal muscle endothelial dysfunction through the activin A–PGC1α axis drives progression of cancer cachexia | Nature Cancer

A microbial amino-acid-conjugated bile acid, tryptophan-cholic acid, improves glucose homeostasis via the orphan receptor MRGPRE | Cell

Expanded surface CD3+ T-cell clones with lymphoma driver somatic mutations accumulate in refractory celiac disease type 1 | Science Translational Medicine

MicroRNA-142-3p shuttling in extracellular vesicles marks regulatory T cell dysfunction in multiple sclerosis | Science Translational Medicine

BRAF oncogenic mutants evade autoinhibition through a common mechanism | Science

The IL-22–oncostatin M axis promotes intestinal inflammation and tumorigenesis | Nature Immunology

Microbiota and chemotherapy response

Bacteroides fragilis promotes chemoresistance in colorectal cancer, and its elimination by phage VA7 restores chemosensitivity | Cell Host & Microbe

Microbiota-derived urocanic acid triggered by tyrosine kinase inhibitors potentiates cancer immunotherapy efficacy | Cell Host & Microbe

Gut microbial metabolite 4-hydroxybenzeneacetic acid drives colorectal cancer progression via accumulation of immunosuppressive PMN-MDSCs | JCI

Drug-induced cardiotoxicity

Cardiomyocyte-localized CCDC25 senses NET DNA to promote doxorubicin cardiotoxicity by activating autophagic flux | Nature Cancer

Topobexin avoids cardiotoxic effects of conventional anthracycline chemotherapeutics by selectively targeting the ATPase domain of the beta isoform of topoisomerase II | Nature Communications

Therapeutic modalities

Base editing of trinucleotide repeats that cause Huntington’s disease and Friedreich’s ataxia reduces somatic repeat expansions in patient cells and in mice | Nature Genetics

Insilico Medicine’s oral ENPP1 inhibitor designed using generative AI as STING modulator in solid tumors | Nature Communications

Developing nanobodies as allosteric molecular chaperones of glucocerebrosidase function | Nature Communications

Single systemic injection of AAV PHP.eB encoding gRNA nested within a synthetic U7 promoter boosts endogenous ADAR RNA editing to 75% in a mouse model of Hurler syndrome | Nature Communications

Systemic delivery of cadherin 17–specific CAR T cells allows effective and safe targeting of colorectal cancer liver metastases | Science Translational Medicine

STING inhibitors encapsulated within a blood–brain barrier-permeable polymeric micelle that scavenges Aβ to target microglia for Alzheimer’s | PNAS

Lemborexant ameliorates tau-mediated sleep loss and neurodegeneration in males in a mouse model of tauopathy | Nature Neuroscience

Bimagrumab blockade of activin type II receptors preserves skeletal muscle mass and enhances fat loss during GLP-1 receptor agonism with semaglutide | Mol Metabolism

Tools and assays

Decoding post-transcriptional regulatory networks by RNA-linked CRISPR screening in human cells | Nature Methods

Lipid nanoparticle (LNP-X; SM-102:DSPC:β-sitosterol:DMG-PEG2000) delivered CRISPRa and Tat mRNAs as HIV latency activators in quiescent CD4+ T cells | Nature Communications

Cells from recessive dystrophic epidermolysis bullosa patients electroporated with CRISPR/Cas9-guide RNA ribonucleoproteins against two large COL7A1 exons (73 and 105) increase expression and deposition of collagen 7 in vivo | Hum Gene Ther

Machine-learning-assisted universal protein activation in living mice | Cell

Targeted apoptotic immune modulator for the treatment of metastatic EGFR-positive solid tumors | PNAS

Startup news

Several startups presented preclinical work at ASCO in Chicago this week:

Startup (location) Preclinical data presented
Great Novel Therapeutics Biotech & Medicals (Taipei City, Taiwan) Combinations of small-molecule histone deacetylase type 1 inhibitor with anti-VEGFR TKI or murine/human PD1 antibody in murine colon cancer model
Secarna Pharmaceuticals (Planegg-Martinsried, Germany) Locked nucleic acid antisense oligonucleotide (ASO) targeting of neuropilin-1 (NRP-1) for combination with checkpoint inhibitors in mouse models of NRP-1-high gastric and breast cancers with poor prognosis
Halda Therapeutics (New Haven, CT) Regulated induced proximity targeting chimera (RIPTAC) targeting full-length androgen receptor and mitotic kinases or epigenetic regulators in metastatic castrate resistant prostate cancer
BlossomHill Therapeutics (San Diego, CA) Reversible tyrosine kinase macrocycle inhibitor targeting active conformation of mutant EGFR tested in cell-derived xenograft or patient-derived xenograft tumor models of non-small cell lung cancer
Merna Therapeutics (Shanghai, China) Sortilin (neurotensin receptor 3)-targeting peptide drug conjugates demonstrated cytotoxicity to platinum-resistant ovarian cancer cell lines
Processa Pharmaceuticals (Hanover, MD) Pro-drug form of active metabolite (SN-38) of irinotecan preferentially accumulates in SW620 colorectal cancer xenograft model
Boundless Bio (San Diego, CA) Testing of CHK1 inhibitor (BBI-355) for esophageal and gastric cancers with EGFR or FGFR2 amplifications on extrachromosomal DNA in preclinical CDX and PDX models together with early phase 1/2 data

At the Yale Innovation Summit, VC keynote announces influx of non-dilutive funding: Reed Jobs of Yosemite outlines new model for supporting life science startups by combining ‘no-stringsattached’ philanthropy and traditional VC funding

University of Texas MD Anderson Cancer Center Cancer Focus Fund eyes a $250 million second fund

Preclinical financings

Date Company (location) Amount (millions) Funding type (lead investors) Therapeutic (lead) focus
May 5, 2025 MetaLead (Basel, Switzerland) CHF1 Seed. UZH Life Sciences Fund (UZH and Novartis Venture Fund) Orally available macrocycle peptides for binding copper and lead for Wilson disease and lead poisoning
May 27, 2025 GlycoEra (Wädenswil, Switzerland) $130 Series B (Novo Holdings) Bifunctional glycan-binding extracellular protein degraders with lead GE8820 targeting pathogenic IgG4 for several autoimmune conditions
May 27, 2025 Hemastatx (Charleroi, Belgium) ND Seed (BaseLaunch, Gemma Frisius Fund and the Butterfly Fund) mAb targeting ADAMTS13 for bleeding disorders of von Willebrand factor
May 29, 2025 Vima Therapeutics (Cambridge, MA) $60 Series A (Atlas, Access Industries and Canaan) Small molecule antagonists of muscarinic cholinergic receptors for dystonia

Preclinical deals

Date Type Payer (location) Payee (location) Upfront payment (millions) Milestones amount (millions) Total (up to millions) Therapeutic Lead Focus
May 27, 2025 R&D collaboration Biogen (Cambridge, MA) City Therapeutics (Cambridge, MA) $16 plus $30 in equity $1,000 $1,460 mRNA cleavage-inducing tiny (CITY) siRNAs against undisclosed target mediating several CNS diseases
May 27, 2025 License Juri Biosciences (San Francisco, CA) EpiMab Biotherapeutics (Shanghai China) ND ND $210 T-cell engager bispecific mAb targeting CD3 and kallikrein-related peptidase to treat metastatic prostate cancer

Stay in touch

We hope you enjoyed this issue of The Needle and hit the button below to receive forthcoming issues into your inbox

If you’re interested in commercializing your science, get in touch. We can help you figure out the next steps for your startup’s translational research program and connect you with the right investor. Follow us on X, BlueSky and LinkedIn. Please send feedback; we’d love to hear from you (info@haystacksci.com).

Until next week,

Juan Carlos and Andy

Subscribe to Haystack Science