WO2025180907A1 - Specific galactoside inhibitor of galectin-1 - Google Patents
Specific galactoside inhibitor of galectin-1Info
- Publication number
- WO2025180907A1 WO2025180907A1 PCT/EP2025/054400 EP2025054400W WO2025180907A1 WO 2025180907 A1 WO2025180907 A1 WO 2025180907A1 EP 2025054400 W EP2025054400 W EP 2025054400W WO 2025180907 A1 WO2025180907 A1 WO 2025180907A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- galectin
- cancer
- formula
- https
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
- A61K31/425—Thiazoles
- A61K31/427—Thiazoles not condensed and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- the present invention relates to a compound of formula (I)
- the present invention also concerns a method of treating a cancer.
- Galectins are a family of lectins that mediate numerous biological functions in both health and disease. Due to their solubility, galectins are present in the extracellular space, cytosol and nucleus, and can act both intra- and extracellularly. Galectins bind beta-galactoside sugars, a common glycan modification on many cell surface receptors, modulating cellular functions by altering receptor-mediated signal transduction pathways (Liu, 2005; Rabinovich and Toscano, 2009). Galectins can also bind intracellular interactors in a carbohydrate-independent manner, with binding partners varying between different galectin family members (Liu and Stowell, 2023).
- the 15 galectin family members (11 in humans) are classified by the number and arrangement of their carbohydrate recognition domains (CRD).
- Prototypical galectins have one, and are found as monomers or homodimers (galectin-1, -2, -5, -7, -10, -11, -13, -14 and - 15); the chimeric galectin-3 has one CRD and an amino-terminal domain which can form oligomers; and the tandem repeat galectins (galectin-4, -6, -8, -9 and -12) contain two distinct CRDs connected by an amino acid linker, and are present as monomers or dimers (Cummings et al., 2009; Sciacchitano et al., 2018; Seyrek, Richter and Lavrik, 2019; Liu and Stowell, 2023).
- Galectin-1 regulates both physiological and disease-related processes, including tissue growth, cellular proliferation, apoptosis, vascularisation, inflammation and carcinogenesis (Perillo et al., 1995; Cousin and Cloninger, 2016; Yu et al., 2023).
- galectin-1 is generally considered immunosuppressive due to its ability to induce apoptosis of effector T cells, antagonise T cell receptor signalling, block T cell-extracellular matrix interactions, and activate regulatory T cell populations (Perillo et al., 1995; Rabinovich G A et aL, 1999; Liu et al., 2009; Cutine et al., 2021).
- Galectin-1 also polarises macrophages to an anti-inflammatory “M2” profile, and can restrict reactive oxygen species generation in neutrophils (Correa et al., 2003; Rodrigues et al., 2019). In a tumour microenvironment, this suppression of T cells in particular may contribute to the persistence of tumours. Galectin-1 is implicated in other hallmarks of cancer, including the promotion of tumour angiogenesis, desmoplasia and hypoxia (Ito et al., 2012; Griffioen and Thijssen, 2014; Kuo and Le, 2014; Martinez-Bosch et al., 2014; Stanley, 2014; van Beijnum et al., 2016).
- galectin-1 specifically in cancer is less well studied, although galectin-1 expression is known to vary in different cancer types, and several analyses have demonstrated correlations between cancers with high galectin-1 expression and overall survival, such as in pancreatic cancer and gastric cancer (Thijssen etal., 2015; Liu et al., 2020).
- the present invention relates in a first aspect to a compound of formula (I)
- normal tissue means mammalian tissue, such as human tissue, in a subject that does not have cancer.
- the cancer is a solid tumor and the solid tumor is reduced by said treatment.
- the compound of formula I further inhibits immune-suppressive cytokines, wherein the cytokines comprise at least IL- 17, IL- 10 and IL-6.
- the compound of formula (I) is 3,4-Dichlorophenyl 3-deoxy- 3-[4-(2-hydroxythiazol-4-yl)-17/-l,2,3-triazol-l-yl]-2-O-methyl-l-thio-a-D-galactopyranoside.
- the cancer is selected from sarcoma, mesothelioma, glioblastoma, carcinosarcoma, adenocarcinoma, adrenocortical cancer, carcinoma and melanoma.
- the cancer is melanoma.
- the cancer is carcinoma.
- the cancer is breast invasive carcinoma.
- the cancer is skin cutaneous melanoma.
- the treatment is by oral administration of the compound of formula (I).
- the compound of formula I is administered in a daily dose from lOmg to 2000mg to a human subject to a human subject.
- the compound of formula I is administered twice daily in a unit dose from 5mg to lOOOmg.
- the compound of formula I is administered in a daily dose from 50mg to 600mg to a human subject.
- the compound of formula I is administered twice daily in a unit dose from 25mg to 300mg.
- the present invention relates to an oral pharmaceutical composition
- an oral pharmaceutical composition comprising the compound of formula I (3,4-Dichlorophenyl 3-deoxy-3-[4-(2-hydroxythiazol-4- yl)-17/-l,2,3-triazol-l-yl]-2-O-methyl-l-thio-a-D-galactopyranoside), and optionally a pharmaceutically acceptable additive.
- the oral pharmaceutical composition of the second aspect is selected from a tablet, a pill, a capsule, granules, a solution, or a suspension.
- the present invention relates to a method for treatment of a cancer having a high expression of galectin-1 comprising administering a therapeutically effective amount of a compound of formula (I)
- GB1908 is a galectin-1 inhibitor that reduces cell surface galectin-1 expression on mouse macrophages. Selectivity of GB1908 for human and mouse galectin-1, galectin-3, galec- tin-9N and galectin-9C (A). Extracellular galectin-1 (B) and galectin-3 (C) were measured by flow cytometry on mouse bone-marrow-derived macrophages, after 2 hours incubation in vitro with a range of doses of GB1908 (B). Data shown as percentage change in mean fluorescence intensity (MFI) from control sample. Each dot represents one mouse; bars show mean +/- SEM. Stars indicate significance using one-way repeated measures ANOVA.
- MFI mean fluorescence intensity
- FIG. 3 Immunomodulatory profiles of galectin-1 and galectin-3 inhibitors GB1908 and GB1211, and Pembrolizumab, in a stromal NSCLC tumour microenvironment model in vitro.
- Primary human immune cells and fibroblasts, and a NSCLC cell line were co-cultured and treated with a dose range of GB1211 (A), GB1908 (B), or 25 pg/mL Pembrolizumab (C, black line; red line is 10 pM GB1908 for comparison).
- Annotated biomarkers indicate at least one concentration of the drug has an effect size >20% and a p value ⁇ 0.01 compared to the control group.
- Prefix “s” indicates soluble biomarker, measured in cell culture supernatant.
- GB1908 reduces IL- 17 and IFN gamma production in stimulated mouse and human T cells.
- T lymphocytes isolated from human whole blood (A, C, E) or mouse spleen (B,D, F) were stimulated with anti-CD3 and anti-CD28, along with a dose range of GB1908 (left column).
- FIG. 5 GB1908 reduces cytokine production in the mouse concanavalin-A model of acute inflammation.
- the compound of formula I is a galectin-1 specific inhibitor. This inhibitor is highly selective for galectin-1 over galectin-3, gal ectin- 9N and galectin-9C ( Figure 1A) and has similar affinities for mouse and human gal ectins (Table 1).
- Galectin-1 expression is known to vary across different cancer types (Cousin and Clon- inger, 2016).
- the Cancer Genome Atlas (TCGA) dataset contains gene expression data from cancers and normal tissue samples from a cohort of 11,000 patients, encompassing 33 different tumour types. To investigate which cancers would benefit from galectin-1 inhibitor therapy, TCGA was mined to identify cancers with the highest expression of galectin-1 (LGALS1) ( Figure 2A). Metadata in TCGA also includes overall and progression-free survival. Correlations between galectin-1 expression and progression-free survival was analysed for two cancer types: breast invasive carcinoma and skin cutaneous melanoma (metastatic). For both cancers, high galectin-1 expression was associated with lower survival probability (Figure 2B, C).
- mice were used in two syngeneic mouse models: breast carcinoma and melanoma.
- Figure 2D the breast carcinoma model
- Figure 2E Treatment with compound of formula I reduced tumour volume.
- galectin-1 is a known regulator of the inflammatory response, particularly T cells, and may also alter the immune response to the tumour.
- the mechanisms of action of the compound of formula I in the context of the tumour immune microenvironment were investigated using the in vitro BioMAP system (Eurofins Discovery, CA, USA). Co-cultures of human peripheral blood mononuclear cells, human primary dermal fibroblasts and the H1299 non-small cell lung cancer (NSCLC) cell line were stimulated with T cell receptor ligands, to create a model of the stromal tumour microenvironment (“StroNSCLC”). Changes in biomarkers were assessed after 48-hour treatment with the compound of formula I.
- GB1211 a galectin-3 inhibitor currently in trials for NSCLC (Aparisi et al., 2023), and pembrolizumab, a PD-1 inhibitor used for the treatment of a variety of cancers, were also assessed for comparison.
- the galectin-3 inhibitor GB1211 ( Figure 3 A) upregulated several biomarkers including IL-6 and IFN gamma (IFNg) at 10 pM.
- the compound of formula I however had a different immunomodulatory profile, inhibiting the expression of numerous immune-suppressive proteins, notably IL- 17, IL- 10 and IL-6 at 10 pM ( Figure 3B).
- the compound of formula I also has a distinct profile in comparison to pembroli- Kursab (Figure 3C), suggesting a differing mechanism of action that may act as a complimentary therapy.
- Figure 3C Prior to in vivo studies, further in vitro investigations were used to determine whether galectin inhibitors result in similar changes in cytokine production in mouse and human T cells. T cells cultured from human whole blood and mouse spleen were stimulated with anti-CD3 and anti-CD28 to induce cytokine production, and co-cultured with a dose range of the compound of formula I for 48 hours.
- the compound of formula I inhibited IL-17 ( Figure 4A-B), IFN gamma (Figure 4C-D) and TNF alpha ( Figure 4E-F) in a dose-dependent manner in both mouse and human T cells.
- This data also suggests that the reduction in cytokines observed using the BioMAP system is at least partially due to the effects of the compound of formula I on T cells.
- Con- A Concanavalin-A
- Elevated inflammatory cytokines from the resulting inflammatory response can be measured in circulation.
- the effects of galectin- 1 inhibition on cytokines were assessed in this model by pre-treating mice orally with the compound of formula I or vehicle (control), one hour prior to con- A injection ( Figure 5 A). Terminal blood samples were collected for analysis 8 hours after con-A injection.
- galectin-1 Several of the signalling pathways regulated by galectin-1 also have downstream effects on the inflammatory response. In both cancer cells and endothelial cells, galectin-1 induces ERK1/2 signalling, which some studies indicate leads to upregulation of NF-KB activity (Dhawan and Richmond, 2002; Huang et al., 2014; Chen etal., 2016). As IL-6 and TNF alpha are upregu- lated by NF-KB, the reduction in these cytokines observed in our in vitro experiments and mouse studies may be due to the suppression of this pathway by the compound of formula I.
- Galectin-1 also induces SDF-1 production from tumour stroma cells such as pancreatic stellate cells; literature suggests SDF-1 signalling through CXCR4 upregulates IL-6 production (Tang et al., 2008; Lu et al., 2009; Chen et al., 2011; Thomas et al., 2015).
- the activation of tissue plasminogen activator (tPA) by galectin-1 upregulates plasminogen conversion to plasmin (Roda et al., 2009).
- IL- 17 is implicated in tumour progression in a wide range of cancer types through various mechanisms, including upregulation of cancer cell migration and metastasis, recruitment of myeloid-derived suppressor cells to induce angiogenesis and suppress antitumour immunity, and increase PD-L1 expression (Zhao etal., 2019; Cui et al., 2021; Liao et al., 2023). Increased PD-L1 promotes resistance to immune checkpoint inhibitor therapy, and blocking IL-17A improved efficacy of anti-PD-1 therapy in mouse models of colorectal cancer (Liu et al., 2021).
- IL-17A In murine myeloma transplant models, pharmacological inhibition or genetic ablation of IL-17A improves outcomes (Vucko- vic et al., 2019).
- IL-17 expression by gamma-delta T cells results in the recruitment of a neutrophil population that supresses cytotoxic-T cells, promoting metastasis (Coffelt et al., 2015; Wu et al., 2020).
- IL-6 is also associated with poorer outcomes in many cancers.
- IL-6 drives the JAK/STAT3 signalling pathway, which suppresses tumour-killing NK cells, effector T cells and dendritic cells, whilst upregulating regulatory T cells and myeloid-derived suppressor cells to create an immunosuppressive environment (Johnson, O’Keefe and Grandis, 2018).
- High IL- 6 also correlates with poor response to atezolizumab (anti-PD-Ll antibody) in advanced breast, kidney and bladder cancers in patients, while in mouse models a combined blockade of PD-L1 and IL-6 receptor improved outcomes compared to atezolizumab alone (Huseni etal., 2023).
- IL-6 production in tumour cells and associated fibroblasts, endothelial cells and dendritic cells is upregulated by IL- 17, and in murine models an IL-6 blockade reduced Thl7 cells and improved outcomes (Wang et al., 2009; Hailemichael et al. , 2022).
- the blockade of immune-suppressive biomarkers by the compound of formula I differs from the galectin-3 inhibitor GB1211, which is showing promise in clinical trials of non-small cell lung cancer (NSCLC) (Aparisi et al., 2023).
- NSCLC non-small cell lung cancer
- Galectin-3 expression is highly correlated with poor response to checkpoint inhibitor therapy in NSCLC (Capalbo et al., 2019).
- Galectin-3 strengthens the interactions between PD-1/PD-L1, reducing the affinity of the immune checkpoint inhibitors pembrolizumab and atezolizumab, an effect reversed by GB1211 (Mabbitt et al., 2023). This unique mechanism of GB1211 in overcoming resistance to checkpoint inhibitor therapy appears specific to galectin-3 inhibition.
- treatment means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder.
- the term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relief the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications.
- the treatment may either be performed in an acute or in a chronic way.
- the patient to be treated is preferably a mammal; in particular, a human being, but it may also include animals, such as dogs, cats, cows, sheep and pigs.
- a therapeutically effective amount of a compound of formula (I) of the present invention as used herein means an amount sufficient to cure, alleviate or partially arrest the clinical manifestations of a given disease and its complications. An amount adequate to accomplish this is defined as "therapeutically effective amount”. Effective amounts for each purpose will depend on the severity of the disease or injury as well as the weight and general state of the subject. It will be understood that determining an appropriate dosage may be achieved using routine experimentation, by constructing a matrix of values and testing different points in the matrix, which is all within the ordinary skills of a trained physician or veterinary.
- the present invention relates to a pharmaceutical composition
- a pharmaceutical composition comprising the compound of formula (I) and optionally a pharmaceutically acceptable additive, such as a carrier or an excipient.
- pharmaceutically acceptable additive is intended without limitation to include carriers, excipients, diluents, adjuvant, colorings, aroma, preservatives etc. that the skilled person would consider using when formulating a compound of the present invention in order to make a pharmaceutical composition.
- the adjuvants, diluents, excipients and/or carriers that may be used in the composition of the invention must be pharmaceutically acceptable in the sense of being compatible with the compound of formula (I) and the other ingredients of the pharmaceutical composition, and not deleterious to the recipient thereof. It is preferred that the compositions shall not contain any material that may cause an adverse reaction, such as an allergic reaction.
- the adjuvants, diluents, excipients and carriers that may be used in the pharmaceutical composition of the invention are well known to a person skilled within the art.
- compositions and particularly pharmaceutical compositions as herein disclosed may, in addition to the compounds herein disclosed, further comprise at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier.
- the pharmaceutical compositions comprise from 1 to 99 % by weight of said at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier and from 1 to 99 % by weight of a compound as herein disclosed.
- the combined amount of the active ingredient and of the pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier may not constitute more than 100% by weight of the composition, particularly the pharmaceutical composition.
- two or more of the compounds as herein disclosed are used in combination for the purposes discussed above.
- composition particularly pharmaceutical composition comprising a compound set forth herein may be adapted for oral, intravenous, topical, intraperitoneal, nasal, buccal, sublingual, or subcutaneous administration, or for administration via the respiratory tract in the form of, for example, an aerosol or an air-suspended fine powder. Therefore, the pharmaceutical composition may be in the form of, for example, tablets, capsules, powders, nanoparticles, crystals, amorphous substances, solutions, transdermal patches or suppositories.
- the compound of formula I as described herein may be prepared as disclosed in W00221/001528 example 9. Herein the compound of formula I is also termed GB1908.
- C57BL/6 mice were purchased from Charles River. C57BL/6 mice were euthanised by approved methods, and femurs and tibias dissected and cleaned. Epiphyses were removed from each bone in turn, and bone marrow cells collected by flushing each bone with a needle and syringe filled with sterile PBS, pooling all marrow cells from each mouse. Cells were filtered through a 100 pm cell strainer and cultured in 6-well cell culture plates in media (DMEM + 10% FBS + 1% penicillin-streptomycin + 2mM L-glutamine + 20 ng/mL M-CSF) at 37 °C with 5% C0 2 . On days 4 and 6 after isolation, cells were washed twice with sterile PBS and fresh media added.
- mice were housed in individually ventilated cages at 22°C with relative humidity 45- 65%, in 12-hour light/dark cycles.
- the murine breast carcinoma cell line 4T1 was cultured in the medium RPMI-1640, 10% fetal calf serum, 1% penicillin/streptomycin, with incubation at 37°C and 5% CO2.
- the murine melanoma adenocarcinoma cell line B16-F10 was cultured in DMEM high Glutamax 1, 10% fetal calf serum, 1% penicillin/streptomycin, with incubation at 37°C and 10% CO2.
- mice in both tumour models were treated according to the same dosing schedule.
- the non-small cell lung cancer line NC1-H1299 and human neonatal dermal fibroblasts were cultured in 96-well plates until confluent.
- GB1211 and the compound of formula I were prepared in DMSO and added to wells. Diluted DMSO-only conditions were used as controls. Each compound was tested in triplicate. After 1 hour, sub-mitotic levels of super antigens were added to wells. These antigens act via the T cell receptor to prime T cells but not drive proliferation. Cells were incubated for 48 hours. Direct ELISA was used to measure cell-associated and cell membrane biomarkers. Soluble biomarkers were measured using HTRF ⁇ detection, bead-based multiplex immunoassay or capture ELISA.
- T cells were isolated from whole blood obtained from donors from the Sygnature Blood Donor panel, using EasySepTM Human T cell Isolation Kit.
- T cells were isolated from the spleens of 6-12-week-old male BALB/c mice, sourced from Charles River UK, using the EasySepTM Mouse T cell Isolation Kit. Isolated cells were resuspended in media (RPMI 1640 with 10% inactivated fetal bovine serum, 1% penicillinstreptomycin, 1% GlutaMax, and 55 pM beta-mercaptoethanol in mouse cell media only).
- 96- well plates were coated with 5 pg/mL anti-CD3 antibody.
- GB1211 and the compound of formula I were prepared in DMSO and added to wells at desired concentrations. Cells were added to wells at a concentration of 100,000 cells/well. Cells were incubated for 30 minutes (37°C, 5% CO2) before anti-CD28 antibody was added to a final concentration of 1 pg/mL. Cells were incubated for a further 48 hours. Supernatants were harvested and stored at -80°C. Human and mouse 5-plex U-Plex kits for analysis of TNFa, IL-2, IL-6, IL-17A and IFNy were obtained from Meso Scale Discovery for cytokine analysis, and plates read using the MESO QuickPlex SQ120MM instrument. Data was analysed using the MSD Workbench software.
- Concanavalin-A mouse models Male C57BL/6 mice received the compound of formula I (30 mg/kg) or GB1211 (10 mg/kg) dissolved in vehicle, or vehicle only (84% PEG + 15% Solutol HS15 + 1% Tween-20) via oral gavage at 10 mL/kg. After 1 hour, mice received con-A (Sigma, C5275; 9 mg/kg, dissolved at 1.5 mg/mL in sterile saline) intravenously via the peripheral vein. Mice were monitored throughout the study and were euthanised prior to the end of the study if moderate clinical signs were breached.
- mice were culled by anaesthetic overdose (intraperitoneal), and blood collected from the vena cava. Blood samples were centrifuged at 400 x g with no brake for 10 minutes, and plasma removed and stored at -20 °C until analysis. Plasma cytokines were measured using BioLegend® LEGENDPlexTM assay (Mouse Inflammation Panel), as per the manufacturer’s protocols. Data was acquired using a BD LSR Fortessa (5 laser) analyser. After acquisition, data was analysed using BioLegend® Qognit software.
Landscapes
- Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Chemical & Material Sciences (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to a compound of formula (I) for use in a method for treatment of a cancer having a higher expression of galectin-1 compared to normal tissue.
Description
SPECIFIC GALACTOSIDE INHIBITOR OF GALECTIN-1
Technical field
The present invention relates to a compound of formula (I)
(i); for use in a method for treatment of a cancer having a higher expression of galectin-1 compared to normal tissue. Further, an oral composition is covered. The present invention also concerns a method of treating a cancer.
Background Art
Galectins are a family of lectins that mediate numerous biological functions in both health and disease. Due to their solubility, galectins are present in the extracellular space, cytosol and nucleus, and can act both intra- and extracellularly. Galectins bind beta-galactoside sugars, a common glycan modification on many cell surface receptors, modulating cellular functions by altering receptor-mediated signal transduction pathways (Liu, 2005; Rabinovich and Toscano, 2009). Galectins can also bind intracellular interactors in a carbohydrate-independent manner, with binding partners varying between different galectin family members (Liu and Stowell, 2023). The 15 galectin family members (11 in humans) are classified by the number and arrangement of their carbohydrate recognition domains (CRD). Prototypical galectins have one, and are found as monomers or homodimers (galectin-1, -2, -5, -7, -10, -11, -13, -14 and - 15); the chimeric galectin-3 has one CRD and an amino-terminal domain which can form oligomers; and the tandem repeat galectins (galectin-4, -6, -8, -9 and -12) contain two distinct CRDs connected by an amino acid linker, and are present as monomers or dimers (Cummings et al., 2009; Sciacchitano et al., 2018; Seyrek, Richter and Lavrik, 2019; Liu and Stowell, 2023).
Galectin-1 regulates both physiological and disease-related processes, including tissue growth, cellular proliferation, apoptosis, vascularisation, inflammation and carcinogenesis (Perillo et al., 1995; Cousin and Cloninger, 2016; Yu et al., 2023). In inflammation, galectin-1 is generally considered immunosuppressive due to its ability to induce apoptosis of effector T cells, antagonise T cell receptor signalling, block T cell-extracellular matrix interactions, and activate regulatory T cell populations (Perillo et al., 1995; Rabinovich G A et aL, 1999; Liu et al., 2009; Cutine et al., 2021). Galectin-1 also polarises macrophages to an anti-inflammatory “M2” profile, and can restrict reactive oxygen species generation in neutrophils (Correa et al., 2003; Rodrigues et al., 2019). In a tumour microenvironment, this suppression of T cells in particular may contribute to the persistence of tumours. Galectin-1 is implicated in other hallmarks of cancer, including the promotion of tumour angiogenesis, desmoplasia and hypoxia (Ito et al., 2012; Griffioen and Thijssen, 2014; Kuo and Le, 2014; Martinez-Bosch et al., 2014; Stanley, 2014; van Beijnum et al., 2016).
Several preclinical and clinical studies using gal ectin-3 inhibitors for cancer and chronic inflammatory diseases are underway, of which some have been shown or are proposed to bind to galectin-1 (Laderach and Compagno, 2023). However, the extent of therapeutic effect from the additional inhibition of galectin-1 with these molecules is difficult to ascertain due to their weak affinity for galectin-1 vs. galectin-3, and the lack of solid pharmacological characterisation. The role of galectin-1 specifically in cancer is less well studied, although galectin-1 expression is known to vary in different cancer types, and several analyses have demonstrated correlations between cancers with high galectin-1 expression and overall survival, such as in pancreatic cancer and gastric cancer (Thijssen etal., 2015; Liu et al., 2020). Several studies indicate the potential efficacy of galectin-1 inhibitors in pre-clinical settings, including human bone and lung cancer cells in vitro, and nerve sheath tumour xenograft models in vivo (Goud et aL, 2019, 2020; Shih et aL, 2019). Blockade of galectin-1 using a monoclonal antibody resulted in reduced tumour size in immunocompetent mice bearing lung carcinoma or melanoma xenograft tumours (Croci etal., 2014). In murine oral carcinoma models, antibody blockade of galectin-1 reduced tumour volume and increased T cell infiltration into the tumour (Nambiar et a/., 2019).
Summary of the invention
The present invention relates in a first aspect to a compound of formula (I)
(i); for use in a method for treatment of a cancer having a higher expression of galectin-1 compared to normal tissue. Here normal tissue means mammalian tissue, such as human tissue, in a subject that does not have cancer.
In an embodiment the cancer is a solid tumor and the solid tumor is reduced by said treatment. In a further embodiment the compound of formula I further inhibits immune-suppressive cytokines, wherein the cytokines comprise at least IL- 17, IL- 10 and IL-6.
In a still further embodiment the compound of formula (I) is 3,4-Dichlorophenyl 3-deoxy- 3-[4-(2-hydroxythiazol-4-yl)-17/-l,2,3-triazol-l-yl]-2-O-methyl-l-thio-a-D-galactopyranoside.
In a further embodiment the cancer is selected from sarcoma, mesothelioma, glioblastoma, carcinosarcoma, adenocarcinoma, adrenocortical cancer, carcinoma and melanoma.
In a still further embodiment the cancer is melanoma.
In a further embodiment the cancer is carcinoma.
In a still further embodiment the cancer is breast invasive carcinoma.
In a further embodiment the cancer is skin cutaneous melanoma.
In a still further embodiment the treatment is by oral administration of the compound of formula (I). Typically, the compound of formula I is administered in a daily dose from lOmg to 2000mg to a human subject to a human subject. In another embodiment the compound of formula I is administered twice daily in a unit dose from 5mg to lOOOmg. In a further embodiment, the compound of formula I is administered in a daily dose from 50mg to 600mg to a human subject. In a still further embodiment the compound of formula I is administered twice daily in a unit dose from 25mg to 300mg.
In a second aspect the present invention relates to an oral pharmaceutical composition comprising the compound of formula I (3,4-Dichlorophenyl 3-deoxy-3-[4-(2-hydroxythiazol-4- yl)-17/-l,2,3-triazol-l-yl]-2-O-methyl-l-thio-a-D-galactopyranoside), and optionally a pharmaceutically acceptable additive.
In a further embodiment the oral pharmaceutical composition of the second aspect is selected from a tablet, a pill, a capsule, granules, a solution, or a suspension.
In a third aspect the present invention relates to a method for treatment of a cancer having a high expression of galectin-1 comprising administering a therapeutically effective amount of a compound of formula (I)
(i); to a mammal, such as a human, in need hereof.
Description of Figures 1-5
Figure 1. GB1908 is a galectin-1 inhibitor that reduces cell surface galectin-1 expression on mouse macrophages. Selectivity of GB1908 for human and mouse galectin-1, galectin-3, galec- tin-9N and galectin-9C (A). Extracellular galectin-1 (B) and galectin-3 (C) were measured by flow cytometry on mouse bone-marrow-derived macrophages, after 2 hours incubation in vitro with a range of doses of GB1908 (B). Data shown as percentage change in mean fluorescence intensity (MFI) from control sample. Each dot represents one mouse; bars show mean +/- SEM. Stars indicate significance using one-way repeated measures ANOVA.
Figure 2. Galectin-1 expression is associated with lower survival rates in breast cancer and melanoma, and inhibition of galectin-1 in vivo reduces tumour size. Cancers with the highest
galectin-1 expression according to TCGA; n=57-1062 biopsies per cancer type (A). High galec- tin-1 (LGALS1) expression correlates with lower survival rates in patients with breast invasive carcinoma (B) and skin cutaneous melanoma (metastatic) (C). Syngeneic mouse models of breast carcinoma (D) and melanoma (E) were used to assess the effect of GB1908 on tumour volume. Stars in C and D indicate statistical significance at each timepoint, using multiple corrected unpaired t tests. Thick lines with error bars show mean +/-SEM; thin lines show data from each mouse.
Figure 3. Immunomodulatory profiles of galectin-1 and galectin-3 inhibitors GB1908 and GB1211, and Pembrolizumab, in a stromal NSCLC tumour microenvironment model in vitro. Primary human immune cells and fibroblasts, and a NSCLC cell line were co-cultured and treated with a dose range of GB1211 (A), GB1908 (B), or 25 pg/mL Pembrolizumab (C, black line; red line is 10 pM GB1908 for comparison). Biomarkers listed on the x axes; changes in each biomarker are shown as a log-transformed ratio for the drug treated sample (n=3 per dose) in comparison to the control treatment group (grey “envelope”, n=6). Annotated biomarkers indicate at least one concentration of the drug has an effect size >20% and a p value < 0.01 compared to the control group. Prefix “s” indicates soluble biomarker, measured in cell culture supernatant.
Figure 4. GB1908 reduces IL- 17 and IFN gamma production in stimulated mouse and human T cells. T lymphocytes isolated from human whole blood (A, C, E) or mouse spleen (B,D, F) were stimulated with anti-CD3 and anti-CD28, along with a dose range of GB1908 (left column). IL- 17 (A-B), IFN gamma (C-D) and TNF alpha (E-F) measured in supernatants after 48 hours. Bars show mean +/- SEM, each dot representing one donor (n=3). Data shown as % change from control (DMSO) treated cells for each donor. Stars indicate significance using repeated measured one-way ANOVA.
Figure 5. GB1908 reduces cytokine production in the mouse concanavalin-A model of acute inflammation. Summary of the study (A). IL-17A, IFN gamma, IL-6 and TNF alpha were
measured in terminal blood samples by LEGENDPlex assay (B-E). Bars show mean +/- SEM, each dot represents one mouse. Stars indicate significance using unpaired t test (vehicle vs GB1908).
Detailed description
The compound of formula I is a galectin-1 specific inhibitor. This inhibitor is highly selective for galectin-1 over galectin-3, gal ectin- 9N and galectin-9C (Figure 1A) and has similar affinities for mouse and human gal ectins (Table 1).
Table 1. Binding affinities of compounds for human and mouse ga- lectins. KD values shown (nM) with SEM if available.
An oral formulation was developed for administration to mice in in vivo studies, making it a useful compound for pre-clinical investigations. Target engagement of galectin-1 was investigated by assessing a reduction in galectin-1 expression on mouse bone marrow-derived macrophages. Treatment with the compound of formula I resulted in a dose-dependent reduction in cell-surface galectin-1 as measured by flow cytometry (Figure IB). Galectin-3 remained unchanged, showing the specificity of this compound (Figure 1C).
Galectin-1 expression is known to vary across different cancer types (Cousin and Clon- inger, 2016). The Cancer Genome Atlas (TCGA) dataset contains gene expression data from cancers and normal tissue samples from a cohort of 11,000 patients, encompassing 33 different tumour types. To investigate which cancers would benefit from galectin-1 inhibitor therapy, TCGA was mined to identify cancers with the highest expression of galectin-1 (LGALS1) (Figure 2A). Metadata in TCGA also includes overall and progression-free survival. Correlations between galectin-1 expression and progression-free survival was analysed for two cancer types:
breast invasive carcinoma and skin cutaneous melanoma (metastatic). For both cancers, high galectin-1 expression was associated with lower survival probability (Figure 2B, C).
The involvement of galectin-1 in tumour progression, along with its association with reduced survival probability, indicates that inhibiting galectin-1 could potentially provide therapeutic advantages in treating these types of cancers. To test this hypothesis, the compound of formula I was used in two syngeneic mouse models: breast carcinoma and melanoma. The mouse breast cancer cell line 4T1, or the mouse melanoma adenocarcinoma cell line B16-F10, were cultured and implanted into the mammary fat pads of mice. After 24 hours, mice were treated with either the compound of formula I (30 mg/kg) or vehicle, and tumour size monitored with callipers. In both the breast carcinoma model (Figure 2D) and in the melanoma model (Figure 2E), treatment with compound of formula I reduced tumour volume.
In addition to directly impacting tumour progression mechanisms, galectin-1 is a known regulator of the inflammatory response, particularly T cells, and may also alter the immune response to the tumour. The mechanisms of action of the compound of formula I in the context of the tumour immune microenvironment were investigated using the in vitro BioMAP system (Eurofins Discovery, CA, USA). Co-cultures of human peripheral blood mononuclear cells, human primary dermal fibroblasts and the H1299 non-small cell lung cancer (NSCLC) cell line were stimulated with T cell receptor ligands, to create a model of the stromal tumour microenvironment (“StroNSCLC”). Changes in biomarkers were assessed after 48-hour treatment with the compound of formula I. Treatment with GB1211, a galectin-3 inhibitor currently in trials for NSCLC (Aparisi et al., 2023), and pembrolizumab, a PD-1 inhibitor used for the treatment of a variety of cancers, were also assessed for comparison. The galectin-3 inhibitor GB1211 (Figure 3 A) upregulated several biomarkers including IL-6 and IFN gamma (IFNg) at 10 pM. The compound of formula I however had a different immunomodulatory profile, inhibiting the expression of numerous immune-suppressive proteins, notably IL- 17, IL- 10 and IL-6 at 10 pM (Figure 3B). The compound of formula I also has a distinct profile in comparison to pembroli- zumab (Figure 3C), suggesting a differing mechanism of action that may act as a complimentary therapy.
Prior to in vivo studies, further in vitro investigations were used to determine whether galectin inhibitors result in similar changes in cytokine production in mouse and human T cells. T cells cultured from human whole blood and mouse spleen were stimulated with anti-CD3 and anti-CD28 to induce cytokine production, and co-cultured with a dose range of the compound of formula I for 48 hours. The compound of formula I inhibited IL-17 (Figure 4A-B), IFN gamma (Figure 4C-D) and TNF alpha (Figure 4E-F) in a dose-dependent manner in both mouse and human T cells. This data also suggests that the reduction in cytokines observed using the BioMAP system is at least partially due to the effects of the compound of formula I on T cells.
As the compound of formula I showed similar effects at inhibiting cytokines in activated T cells from both mouse and human, an in vivo mouse model of T cell-mediated inflammation was utilized. Concanavalin-A (con- A) induces acute liver injury by directly activating T lymphocytes, which are recruited to the liver (Heymann et al., 2015). Elevated inflammatory cytokines from the resulting inflammatory response can be measured in circulation. The effects of galectin- 1 inhibition on cytokines were assessed in this model by pre-treating mice orally with the compound of formula I or vehicle (control), one hour prior to con- A injection (Figure 5 A). Terminal blood samples were collected for analysis 8 hours after con-A injection. All cytokines measured were upregulated in con-A treated mice in comparison to naive mice that did not receive con-A. IL-17A, IFN gamma, IL-6 and TNF alpha were all reduced in the compound of formula I treated mice in comparison to vehicle treated mice (Figure 5B-E).
The ability of the compound of formula I, a high affinity and selective galectin- 1 inhibitor, to reduce tumour growth in mouse models of cancer, and downregulate several immune- suppressive cytokines across in vitro and in vivo studies have been clearly demonstrated. Data mining of literature was used to generate a comprehensive overview of pathways regulated by galectin- 1, in tumour cells and the surrounding stroma, that promote cancer progression. Galectin- 1 in tumour-associated endothelial cells upregulated angiogenesis; in cancer-associated fibroblasts, galectin- 1 promoted desmoplasia. In tumour cells, galectin- 1 regulated pathways involved in epithelial-to-mesenchymal transition, cancer cell migration and invasion, and metastasis.
Several of the signalling pathways regulated by galectin-1 also have downstream effects on the inflammatory response. In both cancer cells and endothelial cells, galectin-1 induces ERK1/2 signalling, which some studies indicate leads to upregulation of NF-KB activity (Dhawan and Richmond, 2002; Huang et al., 2014; Chen etal., 2016). As IL-6 and TNF alpha are upregu- lated by NF-KB, the reduction in these cytokines observed in our in vitro experiments and mouse studies may be due to the suppression of this pathway by the compound of formula I. Galectin-1 also induces SDF-1 production from tumour stroma cells such as pancreatic stellate cells; literature suggests SDF-1 signalling through CXCR4 upregulates IL-6 production (Tang et al., 2008; Lu et al., 2009; Chen et al., 2011; Thomas et al., 2015). The activation of tissue plasminogen activator (tPA) by galectin-1 upregulates plasminogen conversion to plasmin (Roda et al., 2009).
It has been shown that several cytokines known to be immune-suppressive in a tumour microenvironment were reduced with compound of formula I treatment. IL- 17 is implicated in tumour progression in a wide range of cancer types through various mechanisms, including upregulation of cancer cell migration and metastasis, recruitment of myeloid-derived suppressor cells to induce angiogenesis and suppress antitumour immunity, and increase PD-L1 expression (Zhao etal., 2019; Cui et al., 2021; Liao et al., 2023). Increased PD-L1 promotes resistance to immune checkpoint inhibitor therapy, and blocking IL-17A improved efficacy of anti-PD-1 therapy in mouse models of colorectal cancer (Liu et al., 2021). In murine myeloma transplant models, pharmacological inhibition or genetic ablation of IL-17A improves outcomes (Vucko- vic et al., 2019). In breast cancer, IL-17 expression by gamma-delta T cells results in the recruitment of a neutrophil population that supresses cytotoxic-T cells, promoting metastasis (Coffelt et al., 2015; Wu et al., 2020).
Similarly, IL-6 is also associated with poorer outcomes in many cancers. IL-6 drives the JAK/STAT3 signalling pathway, which suppresses tumour-killing NK cells, effector T cells and dendritic cells, whilst upregulating regulatory T cells and myeloid-derived suppressor cells to create an immunosuppressive environment (Johnson, O’Keefe and Grandis, 2018). High IL- 6 also correlates with poor response to atezolizumab (anti-PD-Ll antibody) in advanced breast, kidney and bladder cancers in patients, while in mouse models a combined blockade of PD-L1
and IL-6 receptor improved outcomes compared to atezolizumab alone (Huseni etal., 2023). IL-6 production in tumour cells and associated fibroblasts, endothelial cells and dendritic cells is upregulated by IL- 17, and in murine models an IL-6 blockade reduced Thl7 cells and improved outcomes (Wang et al., 2009; Hailemichael et al. , 2022).
The blockade of immune-suppressive biomarkers by the compound of formula I differs from the galectin-3 inhibitor GB1211, which is showing promise in clinical trials of non-small cell lung cancer (NSCLC) (Aparisi et al., 2023). Galectin-3 expression is highly correlated with poor response to checkpoint inhibitor therapy in NSCLC (Capalbo et al., 2019). Galectin-3 strengthens the interactions between PD-1/PD-L1, reducing the affinity of the immune checkpoint inhibitors pembrolizumab and atezolizumab, an effect reversed by GB1211 (Mabbitt et al., 2023). This unique mechanism of GB1211 in overcoming resistance to checkpoint inhibitor therapy appears specific to galectin-3 inhibition. Additionally, expression of gal ectin- 1 and galectin-3, and correlation with survival, varies widely across different cancers. Research identified breast carcinoma and metastatic skin cutaneous melanoma as cancers with both high galec- tin-1 expression, and a correlation between gal ectin- 1 and poorer survival outcomes, suggesting these cancers may benefit from galectin-1 inhibitor therapy. Consideration of the cancer type in which these drugs are tested may improve the likelihood of successful clinical trials.
The term “treatment” and “treating” as used herein means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder. The term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relief the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications. The treatment may either be performed in an acute or in a chronic way. The patient to be treated is preferably a mammal; in particular, a human being, but it may also include animals, such as dogs, cats, cows, sheep and pigs.
The term "a therapeutically effective amount" of a compound of formula (I) of the present invention as used herein means an amount sufficient to cure, alleviate or partially arrest the clinical manifestations of a given disease and its complications. An amount adequate to accomplish this is defined as "therapeutically effective amount". Effective amounts for each purpose will depend on the severity of the disease or injury as well as the weight and general state of the subject. It will be understood that determining an appropriate dosage may be achieved using routine experimentation, by constructing a matrix of values and testing different points in the matrix, which is all within the ordinary skills of a trained physician or veterinary.
In a still further aspect, the present invention relates to a pharmaceutical composition comprising the compound of formula (I) and optionally a pharmaceutically acceptable additive, such as a carrier or an excipient.
As used herein “pharmaceutically acceptable additive” is intended without limitation to include carriers, excipients, diluents, adjuvant, colorings, aroma, preservatives etc. that the skilled person would consider using when formulating a compound of the present invention in order to make a pharmaceutical composition.
The adjuvants, diluents, excipients and/or carriers that may be used in the composition of the invention must be pharmaceutically acceptable in the sense of being compatible with the compound of formula (I) and the other ingredients of the pharmaceutical composition, and not deleterious to the recipient thereof. It is preferred that the compositions shall not contain any material that may cause an adverse reaction, such as an allergic reaction. The adjuvants, diluents, excipients and carriers that may be used in the pharmaceutical composition of the invention are well known to a person skilled within the art.
As mentioned above, the compositions and particularly pharmaceutical compositions as herein disclosed may, in addition to the compounds herein disclosed, further comprise at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier. In some embodiments, the pharmaceutical compositions comprise from 1 to 99 % by weight of said at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier and from 1 to 99 % by weight of a compound as herein disclosed. The combined amount of the active ingredient and
of the pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier may not constitute more than 100% by weight of the composition, particularly the pharmaceutical composition.
In some embodiments, only one compound as herein disclosed is used for the purposes discussed above.
In some embodiments, two or more of the compounds as herein disclosed are used in combination for the purposes discussed above.
The composition, particularly pharmaceutical composition comprising a compound set forth herein may be adapted for oral, intravenous, topical, intraperitoneal, nasal, buccal, sublingual, or subcutaneous administration, or for administration via the respiratory tract in the form of, for example, an aerosol or an air-suspended fine powder. Therefore, the pharmaceutical composition may be in the form of, for example, tablets, capsules, powders, nanoparticles, crystals, amorphous substances, solutions, transdermal patches or suppositories.
Further embodiments of the process are described in the experimental section herein, and each individual process as well as each starting material constitutes embodiments that may form part of embodiments.
The above embodiments should be seen as referring to any one of the aspects (such as ‘method for treatment’, ‘pharmaceutical composition’, ‘compound for use as a medicament’, or ‘compound for use in a method’) described herein as well as any one of the embodiments described herein unless it is specified that an embodiment relates to a certain aspect or aspects of the present invention.
All references, including publications, patent applications and patents, cited herein are hereby incorporated by reference to the same extent as if each reference was individually and specifically indicated to be incorporated by reference and was set forth in its entirety herein.
All headings and sub-headings are used herein for convenience only and should not be construed as limiting the invention in any way.
Any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
The terms “a” and “an” and “the” and similar referents as used in the context of describing the invention are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms “a” and “an” means “one or more” and is interchangeable with “at least one”.
Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. Unless otherwise stated, all exact values provided herein are representative of corresponding approximate values (e.g., all exact exemplary values provided with respect to a particular factor or measurement can be considered to also pro-vide a corresponding approximate measurement, modified by "about," where appropriate).
All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context.
The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise indicated. No language in the specification should be construed as indicating any element is essential to the practice of the invention unless as much is explicitly stated.
The citation and incorporation of patent documents herein is done for convenience only and does not reflect any view of the validity, patentability and/or enforceability of such patent documents.
The term “and/or” as used herein is intended to mean both alternatives as well as each of the alternatives individually. For instance, the expression “xxx and/or yyy” means “xxx and yyy”; “xxx”; or “yyy”, all three alternatives are subject to individual embodiments.
The description herein of any aspect or embodiment of the invention using terms such as “comprising”, “having”, “including” or “containing” with reference to an element or elements is intended to provide support for a similar aspect or embodiment of the invention that “consists of’, “consists essentially of’, or “substantially comprises” that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition
described herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by context). This invention includes all modifications and equivalents of the subject matter recited in the aspects or claims presented herein to the maximum extent permitted by applicable law.
The present invention is further illustrated by the following examples that, however, are not to be construed as limiting the scope of protection. The features disclosed in the foregoing description and in the following examples may, both separately and in any combination thereof, be material for realizing the invention in diverse forms thereof.
EXPERIMENTAL
The compound of formula I as described herein may be prepared as disclosed in W00221/001528 example 9. Herein the compound of formula I is also termed GB1908.
Another compound 5-Bromopyri din-3 -yl 3-deoxy-3-[4-(3,4,5-trifluorophenyl)-lH- l,2,3-triazol-l-yl]-l-thio-a-D-galactopyranoside and having the structure
may be prepared as disclosed in WO2021/122719. Herein this compound is also termed GB1211.
C57BL/6 mice were purchased from Charles River. C57BL/6 mice were euthanised by approved methods, and femurs and tibias dissected and cleaned. Epiphyses were removed from each bone in turn, and bone marrow cells collected by flushing each bone with a needle and syringe filled with sterile PBS, pooling all marrow cells from each mouse. Cells were filtered through a 100 pm cell strainer and cultured in 6-well cell culture plates in media (DMEM + 10% FBS + 1% penicillin-streptomycin + 2mM L-glutamine + 20 ng/mL M-CSF) at 37 °C with
5% C02. On days 4 and 6 after isolation, cells were washed twice with sterile PBS and fresh media added.
Experiments were carried out on cells after 7 days of culture. Cells were washed and detached from wells using trypsin-EDTA and a cell scraper and resuspended in DMEM-only media. A viable cell count was performed using Trypan Blue, and media volume adjusted to generate cell suspensions of 1 million viable cells per mL. Cells were transferred to 2 mL Eppen- dorfs (500 pL per tube) containing the compound of formula I or diluted DMSO (control). Cells were incubated at 37 °C for 2 hours, with gentle shaking (300 rpm).
After incubation, cells were stained with Zombie-UV viability dye (1 : 1000 in PBS) and incubated at room temperature in the dark for 30 minutes. Cells were centrifuged at 300 x g for 5 minutes and supernatants discarded. Into the residual volume of the tube, 2 pL FC-block was added, and cells gently resuspended and incubated at room temperature for 5 minutes. Directly into the residual volume/Fc block, 1 pL of each antibody was added: CD1 lb (Brilliant Violet 605), F4/80 (AlexaFluor 700), Galectin-3 (FITC), Galectin-1 (PE). Samples were incubated at 4°C for 20 minutes. 1 mL FACS cell staining buffer was added to each sample, and samples centrifuged at 300 x g for 5 minutes. Samples were resuspended in 300 pL FACS cell staining buffer and stored on ice until analysis. Samples were analysed using a BD LSR Fortessa (5 laser) analyser with BD FACSDiva™ software.
Syngeneic mouse cancer models
Mice were housed in individually ventilated cages at 22°C with relative humidity 45- 65%, in 12-hour light/dark cycles.
The murine breast carcinoma cell line 4T1 was cultured in the medium RPMI-1640, 10% fetal calf serum, 1% penicillin/streptomycin, with incubation at 37°C and 5% CO2. The murine melanoma adenocarcinoma cell line B16-F10 was cultured in DMEM high Glutamax 1, 10% fetal calf serum, 1% penicillin/streptomycin, with incubation at 37°C and 10% CO2. When confluent, 10,000 4T1 cells in 100 pL PBS were implanted into the left mammary fat pad of female BALB/c mice (n=16), or 20,000 B16-F10 cells were implanted into female C57BL/6N mice (n=16).
Starting one day after tumour cell implantation, mice in both tumour models were treated according to the same dosing schedule. Vehicle group (n=8) received 5 mL/kg PBS by intraperitoneal dosing, three times, 3-4 days apart. GB1908 group (n=8) received 30 mg/kg compound of formula I in PEG 300 + Solutol HS15 + Tween 20 (ratio 84/15/1) at 10 mL/kg volume by oral gavage, twice daily. Tumour size was assessed twice weekly by calliper measurements and calculated as so: width2 x length/2.
BioMAP StroNSCLC
The non-small cell lung cancer line NC1-H1299 and human neonatal dermal fibroblasts were cultured in 96-well plates until confluent. Primary human peripheral blood mononuclear cells (PBMCs) were pooled from multiple donors (n=3-6) and added to the plates. GB1211 and the compound of formula I were prepared in DMSO and added to wells. Diluted DMSO-only conditions were used as controls. Each compound was tested in triplicate. After 1 hour, sub-mitotic levels of super antigens were added to wells. These antigens act via the T cell receptor to prime T cells but not drive proliferation. Cells were incubated for 48 hours. Direct ELISA was used to measure cell-associated and cell membrane biomarkers. Soluble biomarkers were measured using HTRF© detection, bead-based multiplex immunoassay or capture ELISA.
Human and mouse T cell assays
For human assays, T cells were isolated from whole blood obtained from donors from the Sygnature Blood Donor panel, using EasySep™ Human T cell Isolation Kit. For mouse assays, T cells were isolated from the spleens of 6-12-week-old male BALB/c mice, sourced from Charles River UK, using the EasySep™ Mouse T cell Isolation Kit. Isolated cells were resuspended in media (RPMI 1640 with 10% inactivated fetal bovine serum, 1% penicillinstreptomycin, 1% GlutaMax, and 55 pM beta-mercaptoethanol in mouse cell media only). 96- well plates were coated with 5 pg/mL anti-CD3 antibody. GB1211 and the compound of formula I were prepared in DMSO and added to wells at desired concentrations. Cells were added to wells at a concentration of 100,000 cells/well. Cells were incubated for 30 minutes (37°C, 5% CO2) before anti-CD28 antibody was added to a final concentration of 1 pg/mL. Cells were incubated for a further 48 hours. Supernatants were harvested and stored at -80°C. Human and
mouse 5-plex U-Plex kits for analysis of TNFa, IL-2, IL-6, IL-17A and IFNy were obtained from Meso Scale Discovery for cytokine analysis, and plates read using the MESO QuickPlex SQ120MM instrument. Data was analysed using the MSD Workbench software.
Concanavalin-A mouse models Male C57BL/6 mice received the compound of formula I (30 mg/kg) or GB1211 (10 mg/kg) dissolved in vehicle, or vehicle only (84% PEG + 15% Solutol HS15 + 1% Tween-20) via oral gavage at 10 mL/kg. After 1 hour, mice received con-A (Sigma, C5275; 9 mg/kg, dissolved at 1.5 mg/mL in sterile saline) intravenously via the peripheral vein. Mice were monitored throughout the study and were euthanised prior to the end of the study if moderate clinical signs were breached. At 8 hours post-con- A injection, mice were culled by anaesthetic overdose (intraperitoneal), and blood collected from the vena cava. Blood samples were centrifuged at 400 x g with no brake for 10 minutes, and plasma removed and stored at -20 °C until analysis. Plasma cytokines were measured using BioLegend® LEGENDPlex™ assay (Mouse Inflammation Panel), as per the manufacturer’s protocols. Data was acquired using a BD LSR Fortessa (5 laser) analyser. After acquisition, data was analysed using BioLegend® Qognit software.
References:
Abiko, K. et al. (2015) ‘IFN-y from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer’, British Journal of Cancer, 112(9), pp. 1501-1509. Available at: https://doi.org/10.1038/bjc.2015.101.
Antoon, J.W. et al. (2012) ‘Altered Death Receptor Signaling Promotes Epithelial-to-Mesen- chymal Transition and Acquired Chemoresistance’, Scientific Reports, 2(1), p. 539. Available at: https://doi.org/10.1038/srep00539.
Aparisi, F.D.A. et al. (2023) ‘GALLANT-1: GB1211 galectin-3 (Gal-3) inhibitor plus atezoli- zumab for first line treatment in patients with advanced/metastatic non-small cell lung cancer (NSCLC)’ , Annals of Oncology, 34, pp. S829-S830. Available at: https://doi.org/10.1016/j.an- nonc.2023.09.2491.
Ayers, M. etal. (2017) ‘IFN-y-related mRNA profile predicts clinical response to PD-1 blockade’, The Journal of Clinical Investigation, 127(8), pp. 2930-2940. Available at: https://d01.0rg/l 0.1172/JCI91190. van Beijnum, J.R. et al. (2016) ‘A key role for galectin-1 in sprouting angiogenesis revealed by novel rationally designed antibodies’, International Journal of Cancer, 139(4), pp. 824-835. Available at: https://doi.0rg/https://doi.org/lO.1002/ijc.30131.
Bellucci, R. et al. (2015) ‘Interferon-y-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression’, Oncolmmunology, 4(6), p. el008824. Available at: https://doi.org/10.1080/2162402X.2015.1008824.
Ben-Baruch, A. (2012) ‘The Tumor-Promoting Flow of Cells Into, Within and Out of the Tumor Site: Regulation by the Inflammatory Axis of TNFa and Chemokines’, Cancer Microenvironment, 5(2), pp. 151-164. Available at: https://doi.org/10.1007/sl2307-011-0094-3.
Capalbo, C. et al. (2019) ‘Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy: The Galectin-3 Signature in NSCLCs’, International Journal of Molecular Sciences, 20(7). Available at: https://doi.org/10.3390/ijms20071607.
Chen, B. et al. (2016) ‘ERK-mediated NF-KB activation through ASIC1 in response to acidosis’, Oncogenesis, 5(12), pp. e279-e279. Available at: https://doi.org/10.1038/oncsis.2016.81.
Chen, H.-T. et al. (2011) ‘Stromal cell-derived factor- 1/CXCR4 promotes IL-6 production in human synovial fibroblasts’, Journal of Cellular Biochemistry, 112(4), pp. 1219-1227. Available at: https://doi.Org/https://doi.org/10.1002/jcb.23043.
Coffelt, S.B. et al. (2015) ‘IL-17-producing yb T cells and neutrophils conspire to promote breast cancer metastasis’, Nature, 522(7556), pp. 345-348. Available at: https://doi.org/10.1038/naturel4282.
Correa, S.G. et al. (2003) ‘Opposite effects of galectin-1 on alternative metabolic pathways of L-arginine in resident, inflammatory, and activated macrophages’, Glycobiology, 13(2), pp. 119-128. Available at: https://doi.org/10.1093/glycob/cwg010.
Cousin, J.M. and Cloninger, M. J. (2016) ‘The Role of Galectin-1 in Cancer Progression, and Synthetic Multivalent Systems for the Study of Galectin-1’, International Journal of Molecular Sciences, 17(9). Available at: https://doi.org/10.3390/ijmsl7091566.
Croci, D.O. et al. (2014) ‘Glycosylation-Dependent Lectin-Receptor Interactions Preserve Angiogenesis in Anti-VEGF Refractory Tumors’, Cell, 156(4), pp. 744-758. Available at: https://doi.Org/10.1016/j.cell.2014.01.043.
Cui, G. et al. (2021) ‘Dynamic stromal cellular reaction throughout human colorectal adenomacarcinoma sequence: A role of TH17/IL-17A’, Biomedicine & Pharmacotherapy, 140, p. 111761. Available at: https://doi.Org/https://doi.org/10.1016/j.biopha.2021.111761.
Cummings, R.D. et al. (2009) Essentials of glycobiology . 4th edition, Varki, A., Cummings, RD, Esko, JD, Freeze, HH, Stanley, P., Bertozzi, CR, Hart, GW, Etzler, ME, Eds. 4th edition. Cold Spring Harbor Laboratory Press, 2022.
Cutine, A.M. et al. (2021) ‘Tissue-specific control of galectin-1 -driven circuits during inflammatory responses’, Glycobiology, 31(8), pp. 891-907. Available at: https://doi.org/10.1093/gly- cob/cwab007.
Dhawan, P. and Richmond, A. (2002) ‘A Novel NF-KB-inducing Kinase-MAPK Signaling Pathway Up-regulates NF-KB Activity in Melanoma Cells’, Journal of Biological Chemistry, 277(10), pp. 7920-7928. Available at: https://doi.Org/https://doi.org/10.1074/jbc.M112210200.
Goldman, M. J. et al. (2020) ‘Visualizing and interpreting cancer genomics data via the Xena platform’, Nature Biotechnology, 38(6), pp. 675-678. Available at: https://doi.org/10.1038/s41587-020-0546-8.
Goud, N.S. et al. (2019) ‘Synthesis and biological evaluation of morpholines linked coumarintriazole hybrids as anticancer agents’, Chemical Biology & Drug Design, 94(5), pp. 1919— 1929. Available at: https://doi.Org/https://doi.org/10. I l l 1/cbdd.13578.
Goud, N.S. et al. (2020) ‘Novel benzimidazole-triazole hybrids as apoptosis inducing agents in lung cancer: Design, synthesis, 18F-radiolabeling & galectin-1 inhibition studies’, Bioorganic Chemistry, 102, p. 104125. Available at: https://doi.Org/https://doi.org/10.1016/j.bioorg.2020.104125.
Griffioen, A.W. and Thijssen, V.L. (2014) ‘Galectins in tumor angiogenesis.’, Annals of Translational Medicine, 2(9), p. 90. Available at: https://doi.Org/10.3978/j. issn.2305- 5839.2014.09.01.
Hailemichael, Y. et al. (2022) ‘Interleukin-6 blockade abrogates immunotherapy toxicity and promotes tumor immunity’, Cancer Cell, 40(5), pp. 509-523. e6. Available at: https://doi.Org/https://doi.org/10.1016/j.ccell.2022.04.004.
Heymann, F. etal. (2015) ‘The concanavalin A model of acute hepatitis in mice’, Laboratory Animals, 49(l_suppl), pp. 12-20. Available at: https://doi.org/10.1177/0023677215572841.
Huang, C.-S. et al. (2014) ‘Galectin-1 Upregulates CXCR4 to Promote Tumor Progression and Poor Outcome in Kidney Cancer’, Journal of the American Society of Nephrology, 25(7).
Available at: https://journals.lww.eom/jasn/fulltext/2014/07000/galectin_l_upregu- lates_cxcr4_to_promote_tumor.16.aspx.
Huseni, M.A. et al. (2023) ‘CD8+ T cell-intrinsic IL-6 signaling promotes resistance to anti-
PD-L1 immunotherapy’, Cell Reports Medicine, 4(1). Available at: https://doi.Org/10.1016/j.xcrm.2022.100878.
Ito, K. etal. (2012) ‘Galectin-1 as a potent target for cancer therapy: role in the tumor microenvironment’, Cancer and Metastasis Reviews, 31(3), pp. 763-778. Available at: https://doi.org/10.1007/sl0555-012-9388-2.
Johnson, D.E., O’Keefe, R.A. and Grandis, J.R. (2018) ‘Targeting the IL-6/JAK/STAT3 signalling axis in cancer’, Nature Reviews Clinical Oncology, 15(4), pp. 234-248. Available at: https://doi.Org/10.1038/nrclinonc.2018.8.
Jorgovanovic, D. el al. (2020) ‘Roles of IFN-y in tumor progression and regression: a review’, Biomarker Research, 8(1), p. 49. Available at: https://doi.org/10.1186/s40364-020-00228-x.
Kuo, P. and Le, Q.-T. (2014) ‘Galectin-1 links tumor hypoxia and radiotherapy’, Glycobiology, 24(10), pp. 921-925. Available at: https://doi.org/10.1093/glycob/cwu062.
Laderach, D. J. and Compagno, D. (2023) ‘Inhibition of galectins in cancer: Biological challenges for their clinical application’, Frontiers in Immunology, 13. Available at: https://doi.org/10.3389/fimmu.2022.1104625.
Liao, H. etal. (2023) ‘IL-17A promotes tumorigenesis and upregulates PD-L1 expression in non-small cell lung cancer’, Journal of Translational Medicine, 21(1), p. 828. Available at: https://doi.org/10.1186/sl2967-023-04365-3.
Liu, C. et al. (2021) ‘Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer’, Journal for ImmunoTherapy of Cancer, 9(1). Available at: https://doi.org/10.1136/jitc-2020-001895.
Liu, F.-T. (2005) ‘Regulatory Roles of Galectins in the Immune Response’, International Archives of Allergy and Immunology, 136(4), pp. 385-400. Available at: https://doi.org/10.1159/000084545.
Liu, F.-T. and Stowell, S.R. (2023) ‘The role of galectins in immunity and infection’, Nature Reviews Immunology, 23(8), pp. 479-494. Available at: https://doi.org/10.1038/s41577-022- 00829-7.
Liu, S.D. etal. (2009) ‘Galectin-1 Tunes TCR Binding and Signal Transduction to Regulate CD8 Burst Sizel’, The Journal of Immunology, 182(9), pp. 5283-5295. Available at: https://doi.org/10.4049/jimmunol.0803811.
Liu, Y. et al. (2020) ‘Galectins for Diagnosis and Prognostic Assessment of Human Diseases:
An Overview of Meta- Analyses’, Medical Science Monitor, 26, p. 0. Available at: https://doi.Org/https://dx.doi.org/10.12659/MSM.923901.
1
Lu, D.-Y. etal. (2009) ‘SDF-lalpha up-regulates interleukin-6 through CXCR4, PI3K/Akt, ERK, and NF-kappaB-dependent pathway in microglia’, European Journal of Pharmacology, 613(1), pp. 146-154. Available at: https://doi.Org/https://doi.org/10.1016/j.ejphar.2009.03.001. Mabbitt, J. et al. (2023) ‘Resistance to anti-PD-l/anti-PD-Ll: galectin-3 inhibition with GB1211 reverses galectin-3 -induced blockade of pembrolizumab and atezolizumab binding to PD-1/PD-L1’, Frontiers in Immunology, 14. Available at: https://doi.org/10.3389/fimmu.2023.1250559.
Martinez-Bosch, N. et al. (2014) ‘Galectin-1 Drives Pancreatic Carcinogenesis through Stroma Remodeling and Hedgehog Signaling Activation’, Cancer Research, 74(13), pp. 3512-3524. Available at: https://doi.org/10.1158/0008-5472.CAN-13-3013.
Montfort, A. et al. (2019) ‘The TNF Paradox in Cancer Progression and Immunotherapy’, Frontiers in Immunology, 10. Available at: https://doi.org/10.3389/fimmu.2019.01818.
Nambiar, D.K. et al. (2019) ‘Galectin-1 -driven T cell exclusion in the tumor endothelium promotes immunotherapy resistance’, The Journal of Clinical Investigation, 129(12), pp. 5553- 5567. Available at: https://doi.org/10.1172/JCI129025.
Perillo, N.L. et al. (1995) ‘Apoptosis of T cells mediated by galectin-1 ’, Nature, 378(6558), pp. 736-739. Available at: https://doi.org/10.1038/378736a0.
Powell, I. J. etal. (2021) ‘Pro-inflammatory cytokines and chemokines initiate multiple prostate cancer biologic pathways of cellular proliferation, heterogeneity and metastasis in a racially diverse population and underlie the genetic/biologic mechanism of racial disparity: Update’, Urologic Oncology: Seminars and Original Investigations, 39(1), pp. 34-40. Available at: https://doi.Org/https://doi.org/10.1016/j.urolonc.2020.08.019.
Rabinovich G A et al. (1999) ‘Specific inhibition of T-cell adhesion to extracellular matrix and proinflammatory cytokine secretion by human recombinant galectin-1’, Immunology, 97(1), pp. 100-106. Available at: https://doi.Org/https://doi.org/10.1046/j.1365-2567.1999.00746.x.
Rabinovich, GA. and Toscano, M.A. (2009) ‘Turning “sweet” on immunity: galectin-glycan interactions in immune tolerance and inflammation’, Nature Reviews Immunology, 9(5), pp. 338-352. Available at: https://doi.org/10.1038/nri2536.
Roda, O. et al. (2009) ‘Galectin-1 Is a Novel Functional Receptor for Tissue Plasminogen Activator in Pancreatic Cancer’, Gastroenterology, 136(4), pp. 1379-1390.e5. Available at: https://doi.Org/10.1053/j.gastro.2008.12.039.
Rodrigues, L.C. et al. (2019) ‘Galectin-1 modulation of neutrophil reactive oxygen species production depends on the cell activation state’, Molecular Immunology, 116, pp. 80-89. Available at: https://doi.Org/https://doi.org/10.1016/j.molimm.2019.10.001.
Roth, K. et al. (2019) ‘Dichotomous Role of Plasmin in Regulation of Macrophage Function after Acetaminophen Overdose’, The American Journal of Pathology, 189(10), pp. 1986-2001. Available at: https://doi.Org/10.1016/j.ajpath.2019.07.003.
Sciacchitano, S. et al. (2018) ‘Galectin-3: One Molecule for an Alphabet of Diseases, from A to Z’, International Journal of Molecular Sciences, 19. Available at: https://api.seman- ticscholar. org/CorpusID: 4030426.
Seyrek, K., Richter, M. and Lavrik, I.N. (2019) ‘Decoding the sweet regulation of apoptosis: the role of glycosylation and galectins in apoptotic signaling pathways’, Cell Death & Differentiation, 26(6), pp. 981-993. Available at: https://doi.org/10.1038/s41418-019-0317-6.
Shih, T.-C. etal. (2019) ‘Galectin-1 inhibition induces cell apoptosis through dual suppression of CXCR4 and Ras pathways in human malignant peripheral nerve sheath tumors’, Neuro-On- cology, 21(11), pp. 1389-1400. Available at: https://doi.org/10.1093/neuonc/noz093.
Sorme, P. etal. (2004) ‘Fluorescence polarization as an analytical tool to evaluate galectin-lig- and interactions’, Analytical Biochemistry, 334(1), pp. 36-47. Available at: https://doi.Org/https://doi.org/10.1016/j.ab.2004.06.042.
Spranger, S. et al. (2013) ‘Up-Regulation of PD-L1, IDO, and Tregs in the Melanoma Tumor Microenvironment Is Driven by CD8+ T Cells’, Science Translational Medicine, 5(200), pp. 200ral l6-200ral l6. Available at: https://doi.org/10.1126/scitranslmed.3006504.
Stanley, P. (2014) ‘Galectin-1 Pulls the Strings on VEGFR2’, Cell, 156(4), pp. 625-626. Available at: https://doi.Org/https://doi.org/10.1016/j.cell.2014.01.059.
Sugimoto, M.A. etal. (2017) ‘Plasmin and plasminogen induce macrophage reprogramming and regulate key steps of inflammation resolution via annexin Al’, Blood, 129(21), pp. 2896- 2907. Available at: https://doi.org/10.1182/blood-2016-09-742825.
Tang, C.-H. et al. (2008) ‘Bone-derived SDF-1 stimulates IL-6 release via CXCR4, ERK and NF-KB pathways and promotes osteoclastogenesis in human oral cancer cells’, Carcinogenesis, 29(8), pp. 1483-1492. Available at: https://doi.org/10.1093/carcin/bgn045.
Thijssen, V.L. et al. (2015) ‘Galectin expression in cancer diagnosis and prognosis: A systematic review’, Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 1855(2), pp. 235-247. Available at: https://doi.Org/https://doi.org/10.1016/j.bbcan.2015.03.003.
Thomas, M.N. etal. (2015) ‘SDF-1/CXCR4/CXCR7 is pivotal for vascular smooth muscle cell proliferation and chronic allograft vasculopathy’, Transplant International, 28(12), pp. 1426- 1435. Available at: https://doi.Org/https://doi.org/10. I l l 1/tri.12651.
Vuckovic, S. etal. (2019) ‘Bone marrow transplantation generates T cell-dependent control of myeloma in mice’, The Journal of Clinical Investigation, 129(1), pp. 106-121. Available at: https://doi.org/10.1172/JCI98888.
Wang, L. et al. (2009) ‘IL- 17 can promote tumor growth through an IL-6-Stat3 signaling pathway’, Journal of Experimental Medicine, 206(7), pp. 1457-1464. Available at: https://doi.org/10.1084/jem.20090207.
Wu, L. et al. (2020) TL-17-CXC Chemokine Receptor 2 Axis Facilitates Breast Cancer Progression by Up-Regulating Neutrophil Recruitment’, The American Journal of Pathology, 190(1), pp. 222-233. Available at: https://doi.Org/10.1016/j.ajpath.2019.09.016.
Yu, X. et al. (2023) ‘Galectin-1: A Traditionally Immunosuppressive Protein Displays Context- Dependent Capacities’, International Journal of Molecular Sciences, 24(7). Available at: https://doi.org/10.3390/ijms24076501.
Zetterberg, F.R. etal. (2018) ‘Monosaccharide Derivatives with Low-Nanomolar Lectin Affinity and High Selectivity Based on Combined Fluorine- Ami de, Phenyl-Arginine, Sulfur-n, and Halogen Bond Interactions’, ChemMedChem, 13(2), pp. 133-137. Available at: https://doi.Org/https://doi.org/10.1002/cmdc.201700744.
Zetterberg, F.R. et al. (2022) ‘Discovery and Optimization of the First Highly Effective and Orally Available Galectin-3 Inhibitors for Treatment of Fibrotic Disease’, Journal of Medicinal Chemistry, 65(19), pp. 12626-12638. Available at: https://doi.org/10.1021/acs.jmed- chem.2c00660.
Zhao, J. et al. (2019) ‘The role of interleukin- 17 in tumor development and progression’, Journal of Experimental Medicine, 217(1), p. e20190297. Available at: https://doi.org/10.1084/jem.20190297
Claims
1. A compound of formula (I)
(i); for use in a method for treatment of a cancer having a higher expression of galectin-1 compared to normal tissue.
2. The compound for use of claim 1 wherein the cancer is a solid tumor and the solid tumor is reduced by said treatment.
3. The compound for use of claim 2 wherein the compound of formula I further inhibits immune- suppressive cytokines, wherein the cytokines comprise at least IL- 17, IL- 10 and IL-6.
4. The compound for use of any one of claims 1-3 wherein the compound of formula (I) is 3,4- Dichloropheny 1 3 -deoxy-3 - [4-(2-hydroxythiazol-4-yl)- 1 H- 1 ,2,3 -triazol - 1 -yl] -2-O-methyl- 1 - thio-a-D-galactopyranoside.
5. The compound for use of any one of claims 1-4 wherein the cancer is selected from sarcoma, mesothelioma, glioblastoma, carcinosarcoma, adenocarcinoma, adrenocortical cancer, carcinoma and melanoma.
6. The compound for use of any one of claims 1-5 wherein the cancer is selected from carcinoma and melanoma.
7. The compound for use of any one of claims 1-6 wherein the cancer is selected from breast invasive carcinoma and skin cutaneous melanoma.
8. The compound for use of any one of claims 1-7 wherein the treatment is by oral administration of the compound of formula (I).
9. The compound for use of claim 8 wherein the compound of formula I is administered in a daily dose from lOmg to 2000mg to a human subject.
10. The compound for use of claim 8 or 9 wherein the compound of formula I is administered in twice daily in a unit dose from 5mg to lOOOmg.
11. An oral pharmaceutical composition comprising the compound of formula I of claim 1, and optionally a pharmaceutically acceptable additive.
12. The oral pharmaceutical composition of claim 10 selected from a tablet, a pill, a capsule, granules, a solution, or a suspension.
13. A method for treatment of a cancer having a high expression of galectin-1 comprising administering a therapeutically effective amount of a compound of formula (I)
(i); to a mammal, such as a human, in need hereof.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP24159555.2 | 2024-02-26 | ||
| EP24159555 | 2024-02-26 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025180907A1 true WO2025180907A1 (en) | 2025-09-04 |
Family
ID=90059384
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2025/054400 Pending WO2025180907A1 (en) | 2024-02-26 | 2025-02-19 | Specific galactoside inhibitor of galectin-1 |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025180907A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019137971A1 (en) * | 2018-01-10 | 2019-07-18 | Galecto Biotech Ab | Novel galactoside inhibitor of galectins |
| WO2021001528A1 (en) | 2019-07-03 | 2021-01-07 | Galecto Biotech Ab | Novel galactoside inhibitor of galectins |
| WO2021122719A1 (en) | 2019-12-16 | 2021-06-24 | Galecto Biotech Ab | Large scale process for the preparation of 5-bromopyridin-3-yl-3-deoxy-3-[4-(3,4,5-trifluorophenyl)-1h-1,2,3-triazol-1-yl]- 1-thio-alpha-d-galactopyranoside and a crystalline form thereof |
-
2025
- 2025-02-19 WO PCT/EP2025/054400 patent/WO2025180907A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019137971A1 (en) * | 2018-01-10 | 2019-07-18 | Galecto Biotech Ab | Novel galactoside inhibitor of galectins |
| WO2021001528A1 (en) | 2019-07-03 | 2021-01-07 | Galecto Biotech Ab | Novel galactoside inhibitor of galectins |
| WO2021122719A1 (en) | 2019-12-16 | 2021-06-24 | Galecto Biotech Ab | Large scale process for the preparation of 5-bromopyridin-3-yl-3-deoxy-3-[4-(3,4,5-trifluorophenyl)-1h-1,2,3-triazol-1-yl]- 1-thio-alpha-d-galactopyranoside and a crystalline form thereof |
Non-Patent Citations (68)
| Title |
|---|
| ABIKO, K. ET AL.: "IFN-y from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer", BRITISH JOURNAL OF CANCER, vol. 112, no. 9, 2015, pages 1501 - 1509, XP055411719, DOI: 10.1038/bjc.2015.101 |
| ANTOON, J.W. ET AL.: "Altered Death Receptor Signaling Promotes Epithelial-to-Mesenchymal Transition and Acquired Chemoresistance", SCIENTIFIC REPORTS,, vol. 2, no. 1, 2012, pages 539, Retrieved from the Internet <URL:https://doi.org/10.1038/srep00539> |
| APARISI, F.D.A. ET AL.: "GALLANT-1: GB1211 galectin-3 (Gal-3) inhibitor plus atezolizumab for first line treatment in patients with advanced/metastatic non-small cell lung cancer (NSCLC", ANNALS OF ONCOLOGY,, vol. 34, 2023, pages S829 - S830, Retrieved from the Internet <URL:https://doi.org/10.1016/j.an-nonc.2023.09.2491> |
| AYERS, M. ET AL.: "IFN-y-related mRNA profile predicts clinical response to PD-1 blockade", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 127, no. 8, 2017, pages 2930 - 2940, XP055608325, Retrieved from the Internet <URL:https://doi.org/10.1172/JCI91190> DOI: 10.1172/JCI91190 |
| BELLUCCI, R. ET AL.: "Interferon-y-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression", ONCOIMMUNOLOGY, vol. 4, no. 6, 2015, pages e1008824, XP055305374, Retrieved from the Internet <URL:https://doi.org/10.1080/2162402X.2015.1008824> DOI: 10.1080/2162402X.2015.1008824 |
| BEN-BARUCH, A.: "The Tumor-Promoting Flow of Cells Into, Within and Out of the Tumor Site: Regulation by the Inflammatory Axis of TNFα and Chemokines", CANCER MICROENVIRONMENT, vol. 5, no. 2, 2012, pages 151 - 164, XP035086701, Retrieved from the Internet <URL:https://doi.org/10.1007/s12307-011-0094-3> DOI: 10.1007/s12307-011-0094-3 |
| CAPALBO, C. ET AL.: "Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy: The Galectin-3 Signature in NSCLCs", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 7, 2019, pages 20, Retrieved from the Internet <URL:https://doi.org/10.3390/ijms20071607> |
| CHEN, B. ET AL.: "ERK-mediated NF- B activation through ASIC1 in response to acido-sis", ONCOGENESIS, vol. 5, no. 12, 2016, pages e279 - e279, Retrieved from the Internet <URL:https://doi.org/10.1038/oncsis.2016.81> |
| CHEN, H.-T. ET AL.: "Stromal cell-derived factor-1/CXCR4 promotes IL-6 production in human synovial fibroblasts", JOURNAL OF CELLULAR BIOCHEMISTRY, vol. 112, no. 4, 2011, pages 1219 - 1227, XP055740702, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1002/jcb.23043> DOI: 10.1002/jcb.23043 |
| COFFELT, S.B. ET AL.: "II,-17-producing yδ T cells and neutrophils conspire to promote breast cancer metastasis", NATURE, vol. 522, no. 7556, 2015, pages 345 - 348, Retrieved from the Internet <URL:https://doi.org/10.1038/nature14282> |
| CORREA, S.G. ET AL.: "Opposite effects of galectin-1 on alternative metabolic pathways of L-arginine in resident, inflammatory, and activated macrophages", GLYCOBIOLOGY, vol. 13, no. 2, 2003, pages 119 - 128, Retrieved from the Internet <URL:https://doi.org/10.1093/glycob/cwg010> |
| COUSIN, J.M.CLONINGER, M.J.: "The Role of Galectin-1 in Cancer Progression, and Synthetic Multivalent Systems for the Study of Galectin-1", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 9, 2016, pages 17, Retrieved from the Internet <URL:https://doi.org/10.3390/ijms17091566> |
| CROCI, D.O. ET AL.: "Glycosylation-Dependent Lectin-Receptor Interactions Preserve Angiogenesis in Anti-VEGF Refractory Tumors", CELL, vol. 156, no. 4, 2014, pages 744 - 758, XP055346020, Retrieved from the Internet <URL:https://doi.org/10.1016/j.cell.2014.01.043> DOI: 10.1016/j.cell.2014.01.043 |
| CUI, G. ET AL.: "Dynamic stromal cellular reaction throughout human colorectal adenoma-carcinoma sequence: A role of THI7 IlL-I 7 A", BIOMEDICINE & PHARMACOTHERAPY, vol. 140, 2021, pages 111761, XP086633314, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.biopha.2021.111761> DOI: 10.1016/j.biopha.2021.111761 |
| CUTINE, A.M. ET AL.: "Tissue-specific control of galectin-1-driven circuits during inflammatory responses", GLYCOBIOLOGY, vol. 31, no. 8, 2021, pages 891 - 907, Retrieved from the Internet <URL:https://doi.org/10.1093/gly-cob/cwab007> |
| DHAWAN, P.RICHMOND, A.: "A Novel NF- B-inducing Kinase-MAPK Signaling Pathway Up-regulates NF- B Activity in Melanoma Cells", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 277, no. 10, 2002, pages 7920 - 7928, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1074/jbc.M112210200.> |
| GOLDMAN, M.J. ET AL.: "Visualizing and interpreting cancer genomics data via the Xena platform", NATURE BIOTECHNOLOGY, vol. 38, no. 6, 2020, pages 675 - 678, XP037167652, Retrieved from the Internet <URL:https://doi.org/10.1038/s41587-020-0546-8> DOI: 10.1038/s41587-020-0546-8 |
| GOUD, N. S. ET AL.: "Novel benzimidazole-triazole hybrids as apoptosis inducing agents in lung cancer: Design, synthesis, 18F-radiolabeling & galectin-1 inhibition studies", BIOORGANIC CHEMISTRY, vol. 102, 2020, pages 104125, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.bioorg.2020.104125.> |
| GOUD, N. S. ET AL.: "Synthesis and biological evaluation of morpholines linked coumarin-triazole hybrids as anticancer agents", CHEMICAL BIOLOGY & DRUG DESIGN, vol. 94, no. 5, 2019, pages 1919 - 1929, XP009525370, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1111/cbdd.13578> DOI: 10.1111/cbdd.13578 |
| GRIFFIOEN, A.W.THIJSSEN, V.L: "Galectins in tumor angiogenesis.", ANNALS OF TRANSLATIONAL MEDICINE, vol. 2, no. 9, 2014, pages 90, Retrieved from the Internet <URL:https://doi.org/10.3978/j.issn.2305-5839.2014.09.01> |
| HAILEMICHAEL, Y. ET AL.: "Interleukin-6 blockade abrogates immunotherapy toxicity and promotes tumor immunity", CANCER CELL, vol. 40, no. 5, 2022, pages 509 - 523, XP087049112, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.ccell.2022.04.004> DOI: 10.1016/j.ccell.2022.04.004 |
| HEYMANN, F. ET AL.: "The concanavalin A model of acute hepatitis in mice", LABORATORY ANIMALS, vol. 49, 2015, pages 12 - 20, Retrieved from the Internet <URL:https://doi.org/10.1177/0023677215572841> |
| HUANG, C.-S. ET AL.: "Galectin-1 Upregulates CXCR4 to Promote Tumor Progression and Poor Outcome in Kidney Cancer", JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY, vol. 7, 2014, pages 25, Retrieved from the Internet <URL:https://journals.1ww.com/jasn/fulltext/2014/07000/galectin_1upregulates_cxcrdtopromotetumor.16.aspx> |
| HUSENI, M.A. ET AL.: "CD8+ T cell-intrinsic IL-6 signaling promotes resistance to anti-PD-L1 immunotherapy", CELL REPORTS MEDICINE, vol. 1, 2023, pages 4, Retrieved from the Internet <URL:https://dol.org/10.1016/j.xcrm.2022.100878> |
| ITO, K. ET AL.: "Galectin-1 as a potent target for cancer therapy: role in the tumor microenvironment", CANCER AND METASTASIS REVIEWS, vol. 31, no. 3, 2012, pages 763 - 778, XP035124681, Retrieved from the Internet <URL:https://doi.org/10.1007/s10555-012-9388-2> DOI: 10.1007/s10555-012-9388-2 |
| JOHNSON, D.E.O'KEEFE, R.A.GRANDIS, J.R.: "Targeting the IL-6/JAK/STAT3 signalling axis in cancer", NATURE REVIEWS CLINICAL ONCOLOGY, vol. 15, no. 4, 2018, pages 234 - 248, Retrieved from the Internet <URL:https://doi.org/10.1038/nrclinonc.2018.8> |
| JORGOVANOVIC, D. ET AL.: "Roles of IFN-γ in tumor progression and regression: a review", BIOMARKER RESEARCH, vol. 8, no. 1, 2020, pages 49, XP021282369, Retrieved from the Internet <URL:https://doi.org/10.1186/s40364-020-00228-x> DOI: 10.1186/s40364-020-00228-x |
| KOICHI ITO ET AL: "Galectin-1 as a potent target for cancer therapy: role in the tumor microenvironment", CANCER AND METASTASIS REVIEWS, KLUWER ACADEMIC PUBLISHERS, DO, vol. 31, no. 3 - 4, 17 June 2012 (2012-06-17), pages 763 - 778, XP035124681, ISSN: 1573-7233, DOI: 10.1007/S10555-012-9388-2 * |
| KUO, P.LE, Q.-T.: "Galectin-1 links tumor hypoxia and radiotherapy", GLYCOBIOLOGY, vol. 24, no. 10, 2014, pages 921 - 925, Retrieved from the Internet <URL:https://doi.org/10.1093/glycob/cwu062> |
| LADERACH, D.J.COMPAGNO, D.: "Inhibition of galectins in cancer: Biological challenges for their clinical application", FRONTIERS IN IMMUNOLOGY, 2023, pages 13, Retrieved from the Internet <URL:https://doi.org/10.3389/fimmu.2022.1104625> |
| LIAO, H. ET AL.: "IL-17A promotes tumorigenesis and upregulates PD-L1 expression in non-small cell lung cancer", JOURNAL OF TRANSLATIONAL MEDICINE, vol. 21, no. 1, 2023, pages 828, Retrieved from the Internet <URL:https://doi.org/10.1186/s12967-023-04365-3> |
| LIU, C. ET AL.: "Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 1, 2021, pages 9, Retrieved from the Internet <URL:https://doi.org/10.1136/jitc-2020-001895> |
| LIU, F.-T.: "Regulatory Roles of Galectins in the Immune Response", INTERNATIONAL ARCHIVES OF ALLERGY AND IMMUNOLOGY, vol. 136, no. 4, 2005, pages 385 - 400, Retrieved from the Internet <URL:https://doi.org/10.1159/000084545> |
| LIU, F.-T.STOWELL, S.R.: "The role of galectins in immunity and infection", NATURE REVIEWS IMMUNOLOGY, vol. 23, no. 8, 2023, pages 479 - 494, XP038002346, Retrieved from the Internet <URL:https://doi.org/10.1038/s41577-022-00829-7> DOI: 10.1038/s41577-022-00829-7 |
| LIU, S.D. ET AL.: "Galectin-1 Tunes TCR Binding and Signal Transduction to Regulate CD8 Burst Sizel", THE JOURNAL OF IMMUNOLOGY, vol. 182, no. 9, 2009, pages 5283 - 5295, Retrieved from the Internet <URL:https://doi.org/10.4049/jimmunol.0803811> |
| LIU, Y. ET AL.: "Galectins for Diagnosis and Prognostic Assessment of Human Diseases: An Overview of Meta-Analyses", MEDICAL SCIENCE MONITOR, vol. 26, 2020, Retrieved from the Internet <URL:https://doi.org/https://dx.doi.org/10.12659/MSM.923901> |
| LU, D.-Y. ET AL.: "SDF- I alpha up-regulates interleukin-6 through CXCR4, PI3K/Akt, ERK, and NF-kappaB-dependent pathway in microglia", EUROPEAN JOURNAL OF PHARMACOLOGY, vol. 613, no. 1, 2009, pages 146 - 154, XP026140511, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.ejphar.2009.03.001> DOI: 10.1016/j.ejphar.2009.03.001 |
| MABBITT, J. ET AL.: "Resistance to anti-PD-1/anti-PD-L1: galectin-3 inhibition with GB1211 reverses galectin-3-induced blockade of pembrolizumab and atezolizumab binding to PD-1/PD-L1", FRONTIERS IN IMMUNOLOGY, 2023, pages 14, Retrieved from the Internet <URL:https://doi.org/10.3389/fimmu.2023.1250559> |
| MARTINEZ-BOSCH, N. ET AL.: "Galectin-1 Drives Pancreatic Carcinogenesis through Stroma Remodeling and Hedgehog Signaling Activation", CANCER RESEARCH, vol. 74, no. 13, 2014, pages 3512 - 3524, Retrieved from the Internet <URL:https://doi.org/10.1158/0008-5472.CAN-13-3013> |
| MONTFORT, A. ET AL.: "The TNF Paradox in Cancer Progression and Immunotherapy", FRONTIERS IN IMMUNOLOGY, 2019, pages 10, Retrieved from the Internet <URL:https://doi.org/10.3389/fimmu.2019.01818> |
| NAMBIAR, D.K. ET AL.: "Galectin-1-driven T cell exclusion in the tumor endothelium promotes immunotherapy resistance", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 129, no. 12, 2019, pages 5553 - 5567, Retrieved from the Internet <URL:https://doi.org/10.1172/JCI129025> |
| PERILLO, N.L. ET AL.: "Apoptosis of T cells mediated by galectin-1", NATURE, vol. 378, no. 6558, 1995, pages 736 - 739, XP002034768, Retrieved from the Internet <URL:https://doi.org/10.1038/378736a0> DOI: 10.1038/378736a0 |
| POWELL, I.J. ET AL.: "Pro-inflammatory cytokines and chemokines initiate multiple prostate cancer biologic pathways of cellular proliferation, heterogeneity and metastasis in a racially diverse population and underlie the genetic/biologic mechanism of racial disparity: Update", URO-LOGIC ONCOLOGY: SEMINARS AND ORIGINAL INVESTIGATIONS, vol. 39, no. 1, 2021, pages 34 - 40, XP086394211, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.urolonc.2020.08.019> DOI: 10.1016/j.urolonc.2020.08.019 |
| RABINOVICH G A ET AL.: "Specific inhibition of T-cell adhesion to extracellular matrix and proinflammatory cytokine secretion by human recombinant galectin-1", IMMUNOLOGY, vol. 97, no. 1, 1999, pages 100 - 106, XP001121122, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1046/j.1365-2567.1999.00746.x> DOI: 10.1046/j.1365-2567.1999.00746.x |
| RABINOVICH, G.A.TOSCANO, M.A.: "Turning ''sweet'' on immunity: galectin-glycan interactions in immune tolerance and inflammation", NATURE REVIEWS IMMUNOLOGY, vol. 9, no. 5, 2009, pages 338 - 352, Retrieved from the Internet <URL:https://doi.org/10.1038/nri2536> |
| RODA, O. ET AL.: "Galectin-1 Is a Novel Functional Receptor for Tissue Plasminogen Activator in Pancreatic Cancer", GASTROENTEROLOGY, vol. 136, no. 4, 2009, pages 1379 - 1390, XP026070070, Retrieved from the Internet <URL:https://doi.org/10.1053/j.gastro.2008.12.039> DOI: 10.1053/j.gastro.2008.12.039 |
| RODRIGUES, L. C. ET AL.: "Galectin-1 modulation of neutrophil reactive oxygen species production depends on the cell activation state", MOLECULAR IMMUNOLOGY, vol. 116, 2019, pages 80 - 89, XP085917707, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.molimm.2019.10.001> DOI: 10.1016/j.molimm.2019.10.001 |
| ROTH, K. ET AL.: "Dichotomous Role of Plasmin in Regulation of Macrophage Function after Acetaminophen Overdose", THE AMERICAN JOURNAL OF PATHOLOGY, vol. 189, no. 10, 2019, pages 1986 - 2001, Retrieved from the Internet <URL:https://doi.org/10.1016/j.ajpath.2019.07.003> |
| SCIACCHITANO, S. ET AL.: "Galectin-3: One Molecule for an Alphabet of Diseases, from A to Z", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 2018, pages 19, Retrieved from the Internet <URL:https://api.seman-ticscholar.org/CorpusID:4030426> |
| SEYREK, K.RICHTER, M.LAVRIK, I.N.: "Decoding the sweet regulation of apoptosis: the role of glycosylation and galectins in apoptotic signaling pathways", CELL DEATH & DIFFERENTIATION, vol. 26, no. 6, 2019, pages 981 - 993, XP036906205, Retrieved from the Internet <URL:https://doi.org/10.1038/s41418-019-0317-6> DOI: 10.1038/s41418-019-0317-6 |
| SHIH, T.-C. ET AL.: "Galectin-1 inhibition induces cell apoptosis through dual suppression of CXCR4 and Ras pathways in human malignant peripheral nerve sheath tumors", NEURO-ONCOLOGY, vol. 21, no. 11, 2019, pages 1389 - 1400, Retrieved from the Internet <URL:https://doi.org/10.1093/neuonc/noz093> |
| SORME, P. ET AL.: "Fluorescence polarization as an analytical tool to evaluate galectin-ligand interactions", ANALYTICAL BIOCHEMISTRY, vol. 334, no. 1, 2004, pages 36 - 47, XP004583732, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1O16/j.ab.2004.06.042> DOI: 10.1016/j.ab.2004.06.042 |
| SPRANGER, S. ET AL.: "Up-Regulation of PD-L1, IDO, and Tregs in the Melanoma Tumor Microenvironment Is Driven by CD8+ T Cells", SCIENCE TRANSLATIONAL MEDICINE, vol. 200, 2013, pages 5, Retrieved from the Internet <URL:https://doi.org/10.1126/scitranslmed.3006504> |
| STANLEY, P.: "Galectin-1 Pulls the Strings on VEGFR2", CELL, vol. 156, no. 4, 2014, pages 625 - 626, XP028613910, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.cell.2014.01.059> DOI: 10.1016/j.cell.2014.01.059 |
| SUGIMOTO, M.A. ET AL.: "Plasmin and plasminogen induce macrophage reprogramming and regulate key steps of inflammation resolution via annexin A1", BLOOD, vol. 129, no. 21, 2017, pages 2896 - 2907, Retrieved from the Internet <URL:https://doi.org/10.1182/blood-2016-09-742825> |
| TANG, C.-H. ET AL.: "Bone-derived SDF-1 stimulates IL-6 release via CXCR4, ERK and NF- B pathways and promotes osteoclastogenesis in human oral cancer cells", CARCINOGENESIS, vol. 29, no. 8, 2008, pages 1483 - 1492, Retrieved from the Internet <URL:https://doi.org/10.1093/carcin/bgn045> |
| THIJSSEN VICTOR L ET AL: "Galectin expression in cancer diagnosis and prognosis: A systematic review", BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - REVIEWS ON CANCER, ELSEVIER SCIENCE BV, AMSTERDAM, NL, vol. 1855, no. 2, 25 March 2015 (2015-03-25), pages 235 - 247, XP029575051, ISSN: 0304-419X, DOI: 10.1016/J.BBCAN.2015.03.003 * |
| THIJSSEN, V.L. ET AL.: "Galectin expression in cancer diagnosis and prognosis: A systematic review", BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - REVIEWS ON CANCER, vol. 1855, no. 2, 2015, pages 235 - 247, XP029575051, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1016/j.bbcan.2015.03.003> DOI: 10.1016/j.bbcan.2015.03.003 |
| THOMAS, M.N. ET AL.: "SDF-1/CXCR4/CXCR7 is pivotal for vascular smooth muscle cell proliferation and chronic allograft vasculopathy", TRANSPLANT INTERNATIONAL, vol. 28, no. 12, 2015, pages 1426 - 1435, XP055915365, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1111/tri.12651> DOI: 10.1111/tri.12651 |
| VAN BEIJNUM, J.R. ET AL.: "A key role for galectin-1 in sprouting angiogenesis revealed by novel rationally designed antibodies", INTERNATIONAL JOURNAL OF CANCER, vol. 139, no. 4, 2016, pages 824 - 835, XP071289373, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1002/ijc.30131> DOI: 10.1002/ijc.30131 |
| VUCKOVIC, S. ET AL.: "Bone marrow transplantation generates T cell-dependent control of myeloma in mice", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 129, no. 1, 2019, pages 106 - 121, Retrieved from the Internet <URL:https://dol.org/l0.1172/JC198888> |
| WANG, L. ET AL.: "II,-17 can promote tumor growth through an IL-6-Stat3 signaling pathway", JOURNAL OF EXPERIMENTAL MEDICINE, vol. 206, no. 7, 2009, pages 1457 - 1464, XP002661455, Retrieved from the Internet <URL:https://doi.org/10.1084/jem.20090207> DOI: 10.1084/jem.20090207 |
| WU, L. ET AL.: "II,-17-CXC Chemokine Receptor 2 Axis Facilitates Breast Cancer Progression by Up-Regulating Neutrophil Recruitment", THE AMERICAN JOURNAL OF PATHOLOGY, vol. 190, no. 1, 2020, pages 222 - 233, Retrieved from the Internet <URL:https://doi.org/10.1016/j.ajpath.2019.09.016> |
| YU, X. ET AL.: "Galectin-1: A Traditionally Immunosuppressive Protein Displays Context-Dependent Capacities", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 7, 2023, pages 24, Retrieved from the Internet <URL:https://doi.org/10.3390/ijms24076501> |
| ZETTERBERG FREDRIK R. ET AL: "Discovery of the Selective and Orally Available Galectin-1 Inhibitor GB1908 as a Potential Treatment for Lung Cancer", vol. 67, no. 11, 13 June 2024 (2024-06-13), US, pages 9374 - 9388, XP093192242, ISSN: 0022-2623, Retrieved from the Internet <URL:https://pubs.acs.org/doi/pdf/10.1021/acs.jmedchem.4c00485> DOI: 10.1021/acs.jmedchem.4c00485 * |
| ZETTERBERG, F.R. ET AL.: "Discovery and Optimization of the First Highly Effective and Orally Available Galectin-3 Inhibitors for Treatment of Fibrotic Disease", JOURNAL OF MEDICINAL CHEMISTRY, vol. 65, no. 19, 2022, pages 12626 - 12638, Retrieved from the Internet <URL:https://doi.org/10.1021/acs.jmed-chem.2c00660> |
| ZETTERBERG, F.R. ET AL.: "Monosaccharide Derivatives with Low-Nanomolar Lectin Affinity and High Selectivity Based on Combined Fluorine-Amide, Phenyl-Arginine, Sulfur-n, and Halogen Bond Interactions", CHEMMEDCHEM, vol. 13, no. 2, 2018, pages 133 - 137, XP055646666, Retrieved from the Internet <URL:https://doi.org/https://doi.org/10.1002/cmdc.201700744> DOI: 10.1002/cmdc.201700744 |
| ZHAO, J. ET AL.: "The role of interleukin-17 in tumor development and progression", JOURNAL OF EXPERIMENTAL MEDICINE, vol. 217, no. 1, 2019, pages e20190297 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Herting et al. | Tumour-associated macrophage-derived interleukin-1 mediates glioblastoma-associated cerebral oedema | |
| EP2688594B1 (en) | Compositions and their use in the treatment of cancer | |
| Park et al. | Activation of gene expression in human neutrophils by high mobility group box 1 protein | |
| Amin et al. | Survival with AGS-003, an autologous dendritic cell–based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): phase 2 study results | |
| Incio et al. | PlGF/VEGFR-1 signaling promotes macrophage polarization and accelerated tumor progression in obesity | |
| Zhang et al. | A membrane form of TNF-α presented by exosomes delays T cell activation-induced cell death | |
| Dalva-Aydemir et al. | Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin | |
| Wu et al. | Tumor microenvironment following Gemcitabine treatment favors differentiation of immunosuppressive Ly6Chigh myeloid cells | |
| Malamut et al. | IL-15 triggers an antiapoptotic pathway in human intraepithelial lymphocytes that is a potential new target in celiac disease–associated inflammation and lymphomagenesis | |
| Mota et al. | Post-sepsis state induces tumor-associated macrophage accumulation through CXCR4/CXCL12 and favors tumor progression in mice | |
| Gu et al. | Sunitinib impairs the proliferation and function of human peripheral T cell and prevents T-cell-mediated immune response in mice | |
| Monk et al. | Integrative development of a TLR8 agonist for ovarian cancer chemoimmunotherapy | |
| Salemizadeh Parizi et al. | Myeloid-derived suppressor cells (MDSCs) in brain cancer: challenges and therapeutic strategies | |
| Ghanim et al. | Suppressive effect of insulin infusion on chemokines and chemokine receptors | |
| Chen et al. | Antipancreatic cancer effect of DNT cells and the underlying mechanism | |
| Huang et al. | In vitro and in vivo killing effects of methionine enkephalin on osteosarcoma | |
| WO2025180907A1 (en) | Specific galactoside inhibitor of galectin-1 | |
| Schneider et al. | The past, present, and future of immunotherapy for bladder tumors | |
| Redin et al. | Dasatinib remodels the tumor microenvironment and sensitizes small cell lung cancer to immunotherapy | |
| Tabata et al. | Highly aggressive plasmablastic neoplasms in patients with rheumatoid arthritis treated with methotrexate | |
| Yang et al. | Solannm lyratum extract affected immune response in normal and leukemia murine animal in vivo | |
| Muraduzzaman et al. | Intrinsic Apoptotic Pathway Genes of Circulating Blood Neutrophils Triggered during HIV Infection and Remained Stimulated in ART Patients | |
| Rai | Immune Checkpoint Inhibitor Therapy for Prostate Cancer: Present and Future Prospectives | |
| Walle et al. | Cytokine release syndrome-like serum responses after COVID-19 vaccination are frequent but clinically inapparent in cancer patients under immune checkpoint therapy | |
| He et al. | Inhibiting tumor cell-intrinsic UBA6 by inosine augments tumor immunogenicity |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 25706227 Country of ref document: EP Kind code of ref document: A1 |