Riccardo Vergaro
@drvergaro.bsky.social
6 followers 0 following 150 posts
Resident in Anesthesiology, Intensive care & Pain medicine - IRCCS San Matteo, Pavia
Posts Media Videos Starter Packs
WO2025210264 discloses novel NaPi2b-targeting antibody-drug conjugates (ADCs) using exatecan payloads. Optimized linkers, controlled DAR and improved PK yield high tumor selectivity and efficacy in NaPi2b-overexpressing cancers (ovarian, NSCLC, endometrial, TNBC) with enhanced safety.
[Credit:Merck]
Novel cyclic peptides target C1q globular heads to mimic antibody–C1q binding, enabling activation or inhibition of the classical complement pathway. Small (1–2 kDa), stable, and low-immunogenic, they offer precise, low-cost tools for complement modulation in immune diseases.
[Credit: Bristol Uni]
US20250304679A1 describes bispecific CD79b×CD3 antibodies for treating CD79b⁺ B-cell lymphomas and leukaemias. By linking B-cell-specific CD79b with T-cell CD3, these antibodies enable targeted T-cell-mediated cytotoxicity and efficient elimination of malignant B cells.
[Credit: LTZ Therapeutics]
WO2025199666 hydrophilic carbohydrate-based multivalent linkers for antibody–drug conjugates (ADCs) enabling high, tunable DARs (up to 32) and dual-payload designs. They improve solubility, stability, pharmacokinetics, and efficacy, overcoming aggregation and heterogeneity limits.
[Credit: Canwell]
US20250304691A1 describes novel bispecific T-cell engagers combining a high-affinity soluble TCR that binds the Survivin–HLA complex on tumor cells with a CD3-binding domain to activate cytotoxic T cells. This approach enables selective killing of Survivin-expressing cancer cells.
[Credit: AbbVie]
W02025199464 describes anti-TROP2 antibody-drug conjugates (ADCs) using eribulin, a microtubule inhibitor, as payload. Targeting TROP2—an oncogenic driver of MAPK, PI3K-AKT, Wnt/β-catenin, and EMT pathways—these ADCs selectively kill TROP2⁺ tumors with reduced systemic toxicity.
[Credit: Eisai Co.]
(W02025207909) discloses a radiolabelled single-domain antibody (sdAb, ssHN3) targeting glypican-3 (GPC3) for PET imaging of hepatocellular carcinoma and other GPC3-positive cancers. It enables high-contrast, non-invasive detection with optimal pharmacokinetics and tumour specificity.
[Credit: NIH]
W02025207816: modular T-cell engaging proteins (TEPs) present viral/bacterial peptides on B cells and bind B-cell antigens, redirecting endogenous memory cytotoxic T cells to deplete B cells, plasmablasts and plasma cells—off-the-shelf, no CAR-T, less lymphodepletion/CRS.
[Credit: Cue Biopharma]
US20250295799A1 a novel anti-GPC3 ADCs using camptothecin payloads for potent, selective and durable antitumor activity in HCC. It improves tumor penetration, payload stability and bystander effect versus prior tubulin/DNA-targeting ADCs, showing better efficacy and safety.
[Credit: Zymeworks]
Patent W02025206561 discloses a novel linker enabling stable conjugation of oncolytic viruses to antibodies, overcoming ADC limitations such as instability and inefficient payload delivery. The linker ensures targeted, precise, and durable delivery of virus-based therapeutics.
[Credit: GeneMedicine]
WO2025202647 describes EGFR-targeted ADCs using CDK inhibitor payloads. This dual-action approach blocks EGFR signaling and CDK-mediated proliferation, enhancing tumor selectivity, reducing systemic toxicity, and overcoming resistance in aggressive cancers like TNBC.
[Credit: KCL]
(W02025199243) target two CD3 epitopes and PRAME, enhancing T-cell engagement and tumor selectivity. This design overcomes bispecific TCE limits, reducing off-tumor toxicity and improving activation against low-density or HLA-peptide antigens via optimized immune synapse formation.[Credit:Regeneron]
WO2025199030 describes a novel antibody–drug conjugate combining a GLP-1 receptor agonist with an anti-GIPR antibody. This dual-targeted design enhances metabolic control, prolongs half-life, and reduces obesity, insulin resistance, dyslipidemia, NASH, and type 2 diabetes risk.
[Credit: Amgen]
Engineered microbial transglutaminases (MTGases) enable site-specific antibody–drug conjugation (>90% at lysine K222). Unlike natural MTGases, which modify multiple sites, these variants selectively form amide bonds at defined residues, yielding homogeneous ADCs with consistent DARs and PK profiles.
US20250289861A1 a GLP-1–Fc–IGF-1 fusion protein designed for CNS therapy. IGF-1 enables BBB penetration; GLP-1 confers neuroprotection and metabolic regulation; and the Fc domain extends half-life. The construct ensures sustained CNS delivery and enhanced therapeutic efficacy.
[Credit: ImmunoForge]
Anti-HER3 dual-payload ADCs exploit synergistic mechanisms to overcome EGFR TKI resistance in NSCLC. By enhancing HER3 targeting—especially after osimertinib pretreatment—improve efficacy over monotherapy, expand the therapeutic window and address key unmet needs in resistant disease.[Credit:Suzhou]
WO2025192937 engineered human albumin mutants with optimized pH-dependent FcRn binding. Computational modeling identified key residues, enabling mutations that enhance FcRn interaction at acidic pH while releasing at neutral pH. These variants improve drug t1/2, efficacy and reduce dosing burden.
US20250289889A1 discloses activatable masked anti-CTLA-4 antibodies that conceal binding sites until tumor-local activation. This design preserves anti-tumor efficacy while reducing systemic immune-related toxicities, improving safety, dosing, and synergy with PD-1/PD-L1 inhibitors.
[Credit: Xilio]
Petosemtamab, Merus N.V.’s lead antibody, targets EGFR and LGR5. It blocks EGFR signaling, drives EGFR internalization/degradation via LGR5 binding, and boosts immune killing through ADCC and ADCP. Genmab has shown acquisition interest.
WO2025191498 antiSiglec15 antibodies and related modalities (ADCs, bispecifics, CAR-T). By blocking Siglec15, which drives T-cell suppression and osteoclast activity, these agents restore antitumor immunity, complement PD-1/PD-L1 inhibitors and target bone diseases like osteoporosis.[Credit:Adaptam]
US20250281632A1 non-internalising ADCs targeting LGALS3BP, a stromal/tumour-associated glycoprotein driving angiogenesis and metastasis. These release cytotoxics extracellularly, killing tumour and stromal cells, bypassing internalisation limits and exploiting abundant LGALS3BP. [Credit:MediaPharma]
WO2025194123 discloses bispecific antibodies targeting Muc16/CA125 and NaPi-2b, highly expressed in ovarian and other epithelial cancers. The approach enhances tumour selectivity, limits antigen escape, and supports ADC strategies for potent, targeted therapies against malignancies.
[Credit: Aarvik]
WO2025193454 discloses ADCs linking tumour-targeting antibodies to SMARCA2/4 degraders. This enables selective intracellular degradation of SWI/SNF ATPases, exploiting synthetic lethality in SMARCA-mutant cancers while reducing systemic toxicity versus conventional inhibitors.
[Credit: Prelude Ther]
KIF18A inhibitors disrupt chromosome alignment, causing mitotic arrest and synthetic lethality. Strategies include AMG650 macrocycles (Roche, Insilico), VLS-1488 (Volastra, Phase I), HW221043 (Humanwell), and GenSci122’s rigid tetracyclic scaffold, reflecting diverse design approaches.
WO2025186448 ADCs with N-myristoyltransferase (NMT) inhibitors as payloads for refractory cancers. This novel mechanism bypasses resistance to topo/tubulin ADCs, enhances stability, reduces systemic toxicity, and expands therapeutic options with favourable safety and efficacy.
[Credit: Myricx Bio]