The Needle Issue #20


Welcome to The Needle, a newsletter from Haystack Science to help you navigate the latest translational research, with a roundup of the latest news on preclinical biotech startups from around the world.

In this issue, we look at one of biotech’s hottest areas—multi-specific antibodies. Although the hype has somewhat dissipated since Akeso’s September announcement of disappointing overall survival data from the HARMONi-A study of its bi-specific (PD-1 x VEGF) ivonescimab in lung cancer patients, multi-specific antibodies featured in the top three licensing deals and in the second-largest merger of the year. Financings of preclinical startups continued to tick over, with Yale’s Craig Crews’ latest induced-proximity degrader startup, Quarry Thera, catching the eye. This on the back of J&J’s announcement of a $3 billion buyout of an earlier Crews venture: regulated induced-proximity targeting chimera (RIPTAC) developer Halda Therapeutics. Preclinical deals continued to tick over, with several deals from GSK. As usual, anything we missed in the biotech startup world, let us know (info@haystacksci.com).

Haystack chat

By our count, there are now 15 bi-specific antibodies approved by the US Food and Drug Administration (the last peer-reviewed count from 2024 we found chalked up 13). This year has been a bumper year for bi-specifics — antibodies that recognize two molecular targets. Several of 2025’s largest deals have involved assets in this class, including Genmab’s $8 billion acquisition of Merus in September and Takeda’s $11.4 billion splurge on an anti-Claudin18.2 bi-specific antibody and antibody-drug conjugate (ADC) from Innovent Biologics.

Not only is this trend likely to continue, but we predict that it will expand to encompass tri- and multi-specific antibodies, the development of which is an area of intense research activity. Just a couple of weeks ago, South Korea’s Celltrion clinched a $155 million (biobucks) deal for TriOar’s tri-specific ADCs for cold tumors. And at the SITC meeting last month (which we covered in issue 19) tri-specifics were highlighted by no less than five companies: Nextpoint (B7-H7 x CD3 x TMIGD2), CrossBow (cathepsin G peptide x CD3 x CD28), TJ Biopharma (CDCP1 x CD3 x 4-1BB), Biocytogen (DLL3 x CD3 x 4-1BB) and Radiant Therapeutics (potentially tri-specific/trivalent).

Building an antibody that recognizes three or more targets at the same time is not trivial, though. There are multiple technical, clinical and regulatory hurdles that developers need to overcome before the antibody reaches patients. Why, then, go through the trouble of creating a multi-specific antibody when a bi-specific may show clinical benefit? As it turns out, there are several reasons why a multi-specific antibody may be worth the effort.

First, as tumors often escape by downregulating or mutating a single target epitope, a multi-specific antibody may reduce the likelihood of escape by simultaneously targeting multiple tumor antigens. Second, multi-specifics could increase safety and reduce toxicity of a therapy. For example, a multi-specific antibody can be designed to require co-expression of two or more antigens on the same cell to bind effectively. Healthy cells expressing only one antigen would be spared, thereby reducing off-tumor toxicity. Similarly, targeting multiple mechanisms with a single antibody may reduce the need to use several separate drugs, simplifying dosing and reducing risks for patients. Third, and perhaps most important, a multi-specific antibody can simultaneously block several disease pathways, yielding synergistic effects that a bi-specific might not achieve. In solid tumors, for example, tumor heterogeneity, limited immune-cell infiltration and an immunosuppressive microenvironment often result in therapeutic failure. Multi-specific antibodies could combine tumor targeting, immune-cell recruitment and checkpoint modulation in a single molecule.

Perhaps the best example of this comes from the field of T-cell engagers (TCEs). A tri-specific antibody can incorporate not only tumor-cell binding and CD3 engagement, but also a co-stimulatory domain, such as CD28. This can boost T-cell activation, persistence and potency more than a bi-specific that only binds to CD3.

In this regard, a recent paper in PNAS is an excellent example of the power of the approach. A research team from EvolveImmune Therapeutics reports on the development of EVOLVE, a next-generation TCE that integrates CD3 binding with CD2-mediated co-stimulation to enhance T-cell activation, durability and tumor-killing capacity, while avoiding target-independent toxicity.

Conventional CD3-bi-specific TCEs activate T cells through a stimulation signal but often fail to provide the complementary co-stimulation necessary for sustained effector function. This can result in T-cell dysfunction, reduced persistence and limited clinical durability. To address this, Jeremy Myers and his colleagues systematically compared multiple costimulatory pathways and identified CD2 as a superior target owing to its broad expression on naïve, activated and exhausted CD8⁺ T cells, and its sustained expression within tumor-infiltrating lymphocytes.

The team engineered tri-specific antibodies that fuse a CD58 extracellular domain (the natural CD2 ligand — Lymphocyte Function-Associated Antigen 3;LFA-3) to affinity-tuned CD3 binders within an IgG-like format. They showed that integrated CD2 co-stimulation substantially improves T-cell viability, proliferation, cytokine production and cytotoxicity across tumor types.

When optimizing the molecule, they found that CD3 affinity must be attenuated: high-affinity CD3 domains cause target-independent T-cell activation and cytokine release (superagonism), whereas intermediate-affinity variants retain potent tumor-directed killing with reduced off-target activation.

The EVOLVE tri-specifics outperformed matched bi-specifics targeting HER2, ULBP2, CD20 and B7-H4, with increases up to >50-fold in potency, depending on the target. The optimized tri-specifics also showed superior tumor control in vivo, achieving durable tumor regression in humanized mouse models even after cessation of the treatment.

Even though tri- and multi-specific antibodies could offer clear advantages over bi-specifics, they are not without problems. From the technical standpoint, multi-specifics combine multiple binding specificities and often non-natural architectures. This feature increases complexity at every step from discovery to manufacturing. The assembly of IgG-like multi-specifics can result in heavy/light and heavy/heavy chain mispairing leading to heterogeneous products. Although antibody engineers have come up with strategies to address this issue, each solution adds constraints to developability.

Multi-specific antibodies can also have lower expression, cause more host-cell stress and require more advanced cell-line engineering or multi-vector expression systems. Moreover, downstream purification often needs additional steps to separate mis-paired species. Similarly, multi-specific antibodies are often less stable, more aggregation-prone, and more sensitive to formulation conditions, impacting shelf life and immunogenicity risk.

It is also important to show identity, purity and functional activity for each specificity and for the multi-specific activity (that is, simultaneous binding, cell-bridging). So, establishing robust potency assays is often the greatest challenge. What is a good model system to design a development candidate going after several targets at the same time? With each additional binder, complexity in discovery and development increases.

From the clinical standpoint, although multi-specifics can potentially be safer than bi-specific antibodies, as we mentioned above, other toxicological risks exist.

TCEs have been known to trigger cytokine-release syndrome, neurotoxicity, or unexpected tissue toxicity if targets are expressed on normal tissues. First-in-human dosing strategies are therefore critical. Moreover, multi-specifics may have non-linear pharmacokinetics (target-mediated clearance for each target), and dual-target engagement can alter distribution and half-life; selecting a safe, effective dose requires integrated PK/PD modeling and biomarker strategy.

And the headaches don’t stop there. Efficacy of a multi-specific may depend on co-expression of two or more targets. Stratifying patients may therefore complicate trial enrollment and endpoint definition, not to mention that it may be necessary to develop companion diagnostics (already expensive and complex for conventional monoclonal antibodies). And related to this point, when multiple targets are engaged, it can be hard to know which specificity caused an adverse event, complicating risk–benefit evaluation and mitigation.

Finally, from the regulatory perspective, although expectations are still evolving, agencies expect a pharmacological package that reflects multi-specific mechanisms, particularly with regards to toxicology. Regulators routinely require robust control strategies to ensure product consistency. Again, this is going to be more complicated for multi-specifics because small changes in manufacturing can alter pairing or potency.

Multi-specific antibodies are gaining momentum. They represent a potentially powerful technology, but many questions still surround their development. Success may depend on striking the right balance between choosing the appropriate therapeutic indication, identifying the simplest effective format, heavy upfront developability and analytical work, and early interactions with regulators to align on pre-clinical packages.

Translational papers: Best of the rest

Target biology

GSK3B directs DNA repair choice and determines tumor response to PARP1 inhibition independent of BRCA1 | JCI

Targeting plasticity in the pyrimidine synthesis pathway potentiates macrophage-mediated phagocytosis in pancreatic cancer models | JCI

NPY orchestrates attraction of cancer cells and CHK2-dependent clonogenicity in the metastatic niche | PNAS

Elucidating pathway-selective biased CCKBR agonism for Alzheimer’s disease treatment | Cell

The synaptic ectokinase VLK triggers the EphB2–NMDAR interaction to drive injury-induced pain | Science

A myeloid trisomy 21-associated gene variant is protective from Alzheimer’s disease | Nature Neuroscience

Estrogen Receptor signaling drives immune evasion and immunotherapy resistance in HR+ breast cancer | JCI

Myeloid GPSM1 regulates atherosclerosis progression by governing monocyte and macrophage activation and chemotaxis | PNAS

Neuroplastin-55 is a receptor of Manf and protects against diet-induced obesity by promoting adipose browning | PNAS

Failure of nerve regeneration in mouse models of diabetes is caused by p35-mediated CDK5 hyperactivity | Science Translational Medicine

NSD2 targeting reverses plasticity and drug resistance in prostate cancer | Nature

Inhibition of 15-hydroxy prostaglandin dehydrogenase promotes cartilage regeneration | Science

Proof-of-concept studies

Extensive restoration of forelimb function in primates with spinal cord injury by neural stem cell transplantation | Nature Biotechnology

Structure-guided design of a synthetic bile acid that inhibits Clostridioides difficile TcdB toxin | Nature Microbiology

Intestinal TGR5-targeted silicon-based carrier-deoxycholic acid conjugate particle improves glycemic control in mice and pigs | Science Translational Medicine

Curing autoimmune diabetes in mice with islet and hematopoietic cell transplantation after conditioning with Jasper Therapeutics’ CD117 antibody-based | JCI

Targeting orthotopic and metastatic pancreatic cancer with allogeneic stem cell–engineered mesothelin-redirected CAR-NKT cells | PNAS

Preclinical assessment of two FcγRI-specific antibodies that competitively inhibit immune complex-FcγRI binding to suppress autoimmune responses | Nature Communications

Lymphotoxin alpha eradicates acute myeloid leukemia and simultaneously promotes healthy hematopoiesis in mice | Science Translational Medicine

Insilico Medicine’s bifunctional PROTAC empowered by AI-generation targets PKMYT1 in cancer | Nature Communications

An RNA interference therapeutic potentially achieves functional cure of chronic hepatitis B virus infection | Nature Communications

Cancer immunotherapy

Thrombospondin-1–CD47 signaling contributes to the development of T cell exhaustion in cancer | Nature Immunology

Macrophage-targeted immunocytokine leverages myeloid, T, and NK cell synergy for cancer immunotherapy | Cell

Target antigen–displaying extracellular vesicles boost CAR T cell efficacy in cell and mouse models of neuroblastoma | Science Translational Medicine

CAR-T-cells targeting the glycoprotein GD2 show potent anti-tumor efficacy in high-risk ependymoma models | JCI

IL-9 as a naturally orthogonal cytokine with optimal JAK/STAT signaling for engineered T cell therapy | Immunity

IL-9 signaling redirects CAR T cell fate toward CD8+ memory and CD4+ cycling states, enhancing antitumor efficacy | Immunity

Tumour-specific bioorthogonal synthesis of proteolysis-targeting chimeras and nanoparticles boosts T cell activity | Nature Biomedical Engineering

Platforms, delivery, editing

Base editing of β0-thalassemia mutations as a therapeutic strategy for severe β-hemoglobinopathies | Science Translational Medicine

Prime editing-installed suppressor tRNAs for disease-agnostic genome editing | Nature

Roche’s approach to expediting hit-to-lead progression in drug discovery through reaction prediction and multi-dimensional optimization | Nature Communications

Startup news

Atlas Venture’s Year in Review video is out. As usual, it’s chockfull of detailed information about current state of pharma and biotech. To whet your appetite, these graphs show that biopharma’s share of venture funding in the US has markedly declined since the COVID-era peak, mirroring a reduction in the number of biotech investors.

Over the other side of the pond, two high-profile European venture-capital firms announced large funds for early-stage companies:

Medicxi Announces €500 Million Fund V

Sofinnova Partners closes €650M ($750M) Capital XI, greatly exceeding initial target to back early-stage healthcare deals

Elsewhere, mega-fund Flagship Pioneering—which distributes close to ~500 million every year and now has 40+ active biotech ventures within its ecosystem (akin to a small country!)—announced strategic partnerships in Saudi Arabia and the UK:

King Abdullah International Medical Research Center and Flagship Pioneering Announce Strategic Partnership to Expand Clinical Research of Novel Medicines in Saudi Arabia

Flagship Pioneering and the Francis Crick Institute Announce Collaboration to Advance Life Sciences Innovation

It also announced a second fund-wide big pharma deal with GSK (following 2023’s deal with Pfizer):

GSK enters research collaboration Flagship Pioneering’s Quotient Therapeutics and Profound Therapeutics

Elsewhere, large-cap companies are investing in Philadelphia and ‘Cellicon valley’:

Thermo Fisher Scientific Opens East Coast Collaboration Center at BioLabs for Advanced Therapeutics to Help Accelerate Cell and Gene Therapy Development

Lilly plans fourth US Gateway Labs in Philadelphia to spur biotech startups

And Abbvie is reaching across the border:

AbbVie and adMare BioInnovations Foster Life Sciences Innovation with the Launch of the AbbVie Biotech Innovators Award in Quebec for oncology, neuroscience, eye care and aesthetics. Submissions close Feb 20 2026

While government funding is dwindling, ARPA-H announced a program to invest in databases detailing mental health readouts for neuromodulatory drugs:

ARPA-H solicits drug/digital medicine developers for effort to gather multimodal/longitudinal psychological, social, digital and biological data for data repository to aid marker and trial design for mental health treatments

Another funder, The Wellcome Trust, has announced a partnership with the UK’s Academy of Medical Sciences to support UK translational researchers:

Welcome-Academy five-year agreement disbursing £27.3 million to UK researchers, part of which will co-fund Academy’s ‘Future Leaders in Innovation, Enterprise and Research’ (FLIER) program

Meanwhile, National Taiwan University has followed the trend of universities starting their own funds:

National Taiwan University launches 30 million fund to kickstart local innovation

Hundreds of biotech leaders sign letter to FDA emphasizing importance of decision-making consistency, transparency and speed to support industry innovation:

No Patient Left Behind sends letter to FDA Commissioner Marty Makary stating need for certainty and predictable regulatory decisions

Preclinical financings

Preclinical deals

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 The Needle