US20220373552A1 - Methods for detecting and treating cholangiocarcinoma - Google Patents

Methods for detecting and treating cholangiocarcinoma Download PDF

Info

Publication number
US20220373552A1
US20220373552A1 US17/238,431 US202117238431A US2022373552A1 US 20220373552 A1 US20220373552 A1 US 20220373552A1 US 202117238431 A US202117238431 A US 202117238431A US 2022373552 A1 US2022373552 A1 US 2022373552A1
Authority
US
United States
Prior art keywords
cholangiocarcinoma
compound
cca
biological sample
blood
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.)
Granted
Application number
US17/238,431
Other versions
US11506664B1 (en
Inventor
Shang-Cheng Hung
Gwo-Bin Lee
Priya Gopinathan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Tsing Hua University NTHU
Academia Sinica
Original Assignee
National Tsing Hua University NTHU
Academia Sinica
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by National Tsing Hua University NTHU, Academia Sinica filed Critical National Tsing Hua University NTHU
Priority to US17/238,431 priority Critical patent/US11506664B1/en
Application granted granted Critical
Publication of US11506664B1 publication Critical patent/US11506664B1/en
Publication of US20220373552A1 publication Critical patent/US20220373552A1/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0006Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid

Definitions

  • the present disclosure in general relates to compounds and methods for the detection of circulating cancerous cells in a biological sample.
  • Cholangiocarcinoma is the second most common primary malignant liver cancer occurring at the bile ducts which drain bile juice from the liver into the small intestine. It is typically characterized by late-stage diagnosis, poor prognosis, high recurrence and frequent metastasis.
  • imaging techniques including ultrasonography, magnetic resonance imaging and computed tomography, and pathological diagnoses by forceps biopsy or brush cytology are the main tools applied to detect CCA. These methods are limited by tumor sizes, localization and lesions so that they are unable to discover CCA tumor at the early stage.
  • the most common tumor biomarkers used to diagnose CCA are carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA). However, the sensitivity and specificity of CA19-9 and CEA are still not satisfactory in clinical applications.
  • Circulating tumor cells have been suggested to play a role in the metastatic spread of carcinomas, accordingly, they may serve as markers for the detection of tumors, particularly, those that metastasized.
  • CTCs Circulating tumor cells
  • their application in the clinical assessment of CCA remains inclusive. Quantification of a relatively low number of CTCs against a background of millions of blood cells has thwarted progress in CCA diagnosis, and affinity-based isolation techniques using CTC-specific antigens such as EpCAM, EphB4, HER2, EGFR, CEA and etc failed to produce results suffice for clinical applications.
  • Inventors of the present invention unexpectedly identify novel octasaccharides that bind to the surface of cholangio-cancerous cells, thus these octasaccharides are useful for detecting cholangio-cancerous cells in a biological sample of a subject, whom is at risk of developing cholangiocarcinoma (CCA) or has CCA.
  • CCA cholangiocarcinoma
  • the first aspect of the present disclosure is directed to a compound of formula (I),
  • R 1 and R 2 are independently H, or —SO 3 M; and M is a monovalent cation selected from the group consisting of sodium ion, potassium ion, lithium ion or ammonium ion.
  • R 1 and R 2 are independently —SO 3 M, and M is the sodium ion.
  • the second aspect of the present disclosure is directed to a method of treating and detecting circulating cholangio-cancerous cells from a biological sample of a subject suspected of having CCA.
  • the method includes the steps of,
  • the compound of formula (I) is coupled to streptavidin, which is conjugated to the outer surface of a magnetic bead.
  • R 1 and R 2 are independently —SO 3 M, and M is the sodium ion.
  • the biological sample suitable for use in the present method include, but are not limited to, blood, plasma, serum, urine, sputum, saliva, tissue sample, biopsy, and tissue lysate.
  • the biological sample is the blood from the subject.
  • the subject has stage III or IV CCA, or metastatic CCA.
  • the method further comprises, before the step (a), pre-treating the blood with a lysis buffer, so as to lyse red blood cells therein.
  • the method comprises administering to the subject an effective amount of the S-1 to ameliorate symptoms associated with the CCA, in which the S-1 is a combination of tegafur, 5-chloro-2,4-dihydropyrimidine (CDHP), and potassium oxonate.
  • the method comprises administering to the subject an effective amount of a combination of gemcitabine and cisplatin to ameliorate symptoms associated with the CCA.
  • the method comprises administering to the subject an effective amount of a combination of S-1, leucovorin, oxaliplatin, and gemcitabine to ameliorate symptoms associated with the CCA.
  • the method further comprises administering to the subject a radiotherapy in addition to the administration of S-1 to ameliorate symptoms associated with the CCA.
  • FIG. 1A Schematic diagrams of a microfluidic chip.
  • the chip features all layers from L 1 to L 5 representing liquid channel layer, air control layer, liquid channel layer, thin PDMS layer and double-sided tape, respectively. Detail structure of layers L 1 , L 2 and L 3 are shown where (i) represents inlets for lysed blood, (ii) represents pellet collection area, and (iii) represents outlets for waste collection. (I) represents the blood treatment unit, and (II) represents the immunofluorescence staining unit.
  • the 13 connections to electromagnetic valves (EMVs) for pneumatic control of the chip are shown as 1 to 13.
  • the 8 microchambers/reagent storing units are labeled as C 1 to C 8 .
  • C 1 cell pellet collection unit
  • C 2 waste collection unit
  • C 3 magnetic bead storage unit
  • C 4 and C 5 1 ⁇ phosphate-buffered saline (PBS) storage unit
  • C 6 the storage unit for secondary antibody (i.e., DyLight 488 and CD45-PE first and later for DAPI)
  • C 7 waste collection unit
  • C 8 anti-CK17 primary antibody storage unit.
  • FIG. 1B Photograph of the microfluidic chip of FIG. 1A .
  • FIG. 2 Photographs depicting immunofluorescence staining of CTC clusters captured by MB-SCH45 or MB-anti-EpCAM. Negative depletion with anti-CD45 Ab and positive enrichment with MB-SCH45 (top 16 panels) and MB-anti-EpCAM (lower 20 panels) for both representative stage III and IV samples. The bottom 4 panels show the CTC culsters after positive enrichment with MB-anti-EpCAM.
  • FIG. 3 The predictive significance of CTC counts. The correlation between CTC count and clinical response to chemotherapy in 5 advanced or metastatic CCA patients was tested. Panels (A) to (E) represent patients A to E. Subpanels I-III in each panel represent normalized CTC counts isolated using MB-SCH45, MB-anti-EpCAM and FACS, respectively. Subpanel IV represents the normalized tumor size using radiographic imaging.
  • the term “patient” or “subject” may be used interchangeably in the present disclosure, and refers to any animal.
  • the animal can be a human subject, or a non-human subject.
  • the subject may be a human, but can also be a mammal in need of veterinary treatment, e.g., domestic animals or game animals, farm animals, and laboratory animals (e.g., rats, mice, guinea pigs, primates, and the like).
  • veterinary treatment e.g., domestic animals or game animals, farm animals, and laboratory animals (e.g., rats, mice, guinea pigs, primates, and the like).
  • Usually the animal is a non-human mammal, such as a non-human primate.
  • Non-human primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus or Pan.
  • Domestic animals and game animals include cows, horses, pigs, sheep, deer, bison, buffalo, mink, felines (e.g., domestic cats, canines (e.g., dogs)), wolf and fox, avian species (e.g., chicken, turkey, and ostrich), and fish (e.g., trout, catfish, and salmon).
  • felines e.g., domestic cats, canines (e.g., dogs)
  • wolf and fox e.g., chicken, turkey, and ostrich
  • fish e.g., trout, catfish, and salmon.
  • contacting is used herein with respect to a cell (e.g., a cell in a biological sample) and refers to any mode of delivery or “administration” of an agent either to cells or the biological sample, in which the agent (e.g., a compound of the present disclosure or a magnet bead pre-coated with the compound of the present disclosure) is brought into contact with one or more cells in sufficient amount to achieve affinity binding therebetween.
  • agent e.g., a compound of the present disclosure or a magnet bead pre-coated with the compound of the present disclosure
  • aspects of the present disclosure relate to the finding that a novel octasaccharide is useful as a biomarker for the identification and/or detection of cancerous cells in a biological sample.
  • the novel octasaccharide is described herein.
  • the present invention relates to a compound of formula (I):
  • R 1 and R 2 are independently H, or —SO 3 M; and M is a monovalent cation selected from the group consisting of sodium ion, potassium ion, lithium ion or ammonium ion.
  • R 1 and R 2 are independently —SO 3 M, and M is the sodium ion.
  • the compound of the present disclosure may be prepared in accordance with procedures described in the working examples. All stereoisomers of the present compounds, such as those which may exist due to asymmetric carbons on the R substituents of the compound of formula (I) including enantiomeric and diastereomeric forms, are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the compound of formula (I) may bind to surfaces of cancerous cells, particularly cancers originated from cholangio bile duct. Accordingly, the present disclosure thus encompasses a method of identifying or detecting cholangio-cancerous cells from a biological sample of a subject suspected of having cholangiocarcinoma (CCA), as well as a method of treating the subject based on the detection of cholangio-cancerous cells in the biological sample.
  • CCA cholangiocarcinoma
  • the present method includes contacting a biological sample of the subject with a compound of formula (I), in which a binding between the compound of formula (I) and the biological sample indicates the presence of cholangio-cancerous cells in the biological sample.
  • the blood sample is pre-treated with a lysis buffer, which will result in the lysis of red blood cells in the blood sample, prior to commencing the present method (i.e., prior to contacting the blood with magnetic beads).
  • the biological sample suitable for use in the present method include but are not limited to, blood, plasma, serum, urine, sputum, saliva, tissue sample, biopsy, and tissue lysate.
  • the biological sample is blood.
  • the method comprises mixing a blood sample of the subject with the present compound.
  • the blood sample does contain cholangio-cancer cells, they will automatically bind with the compound of formula (I) and form a complex, which may be detected via any suitable means, such as enzyme-linked immunosorbent assay (ELISA), immunofluorescent staining and the like; and the presence of the complex is an indication that the subject is positive for CCA.
  • ELISA enzyme-linked immunosorbent assay
  • the blood sample does not contain cholangio-cancer cells, there will not be a binding between the blood sample and the compound of formula (I), and the absence of the binding is an indication that the subject is negative for CCA.
  • the compound of formula (I) is coated on the surface of a magnetic bead via coupling to streptavidin pre-conjugated on the outer surface of the magnetic bead.
  • the magnetic beads having the compound of formula (I) coated thereon would be able to capture the cholangio-cancerous cells present in the biological sample, and the magnetic beads having cholangio-cancerous cells captured thereon may be allocated and harvested via use of a magnet.
  • suitable treatment e.g., radiotherapy, chemotherapy and the like
  • suitable treatment may then be administered to the subject to ameliorate symptoms associated with the CCA.
  • the method therefore includes administering an effective amount of a chemotherapeutic agent to the subject.
  • chemotherapeutic agent suitable for use in the present method include, but are not limited to, S-1, leucovorin, oxaliplatin, gemcitabine, cisplatin, and a combination thereof.
  • an effective amount of the S-1 is administered to the subject to ameliorate symptoms associated with the CCA, in which the S-1 is a combination of tegafur, 5-chloro-2,4-dihydropyrimidine (CDHP), and potassium oxonate.
  • a radiotherapy is administered to the subject before, concurrently with, or after the administration of the S-1.
  • a combination of gemcitabine and cisplatin is administered to the subject to ameliorate symptoms associated with the CCA.
  • a combination of S-1, leucovorin, oxaliplatin, and gemcitabine is administered to the subject to ameliorate symptoms associated with the CCA.
  • the subject has stage III or IV CCA, or metastatic CCA.
  • DMI water deionized water
  • the experimental procedure involved four steps: (1) RBC lysis and isolation of pellet, (2) negative depletion for the removal of WBCs, (3) positive magnetic enrichment, and 4) immunofluorescence staining.
  • RBC were lysed by adding 9 mL of 1 ⁇ RBC lysis buffer (Biolegend, USA) to 1 mL of peripheral blood and incubating at room temperature with gentle mixing on a rotary shaker (TKS ROCKER-S-01, Lab666, Taiwan) for 5 min. Then the sample was dispensed into the topmost PDMS layer of the chip through the inlets provided ( FIG. 1A ). The microfluidic chip was then spun as described above and placed on a PMMA plate with a groove such that the microfluidic chip could be placed inside it. It was then placed on a spin-coater (M and R Nanotechnology Co.
  • M and R Nanotechnology Co M and R Nanotechnology Co.
  • a negative depletion strategy reliant on the removal of leucocytes via their binding to anti-CD45 Ab bound to magnetic beads was employed herein on-chip.
  • the cell-bead complexes were then attracted to a magnet to remove the bound leukocytes from the lysed blood samples.
  • the magnetic bead-CTC complexes were then collected, washed, and transferred to the immunofluorescence staining unit of the chip for further analysis.
  • the bead-cell complexes were suspended in 10 ⁇ L of 1 ⁇ PBS and collected through the waste collection chamber. They were transferred onto the numbered microscopic slide and ProLong Gold Antifade Reagent (Invitrogen) was used to embed the cell-bead complexes.
  • the numbered microscope slide was fabricated using an SU-8 standard photolithography process to facilitate CTC counting. Concisely, a 30 ⁇ m thick layer of SU-8 3035 (MicroChem, USA) was spin-coated on a glass microscope slide. A soft-baking process for 10 min was performed and the SU-8 microstructures were patterned at a dosage of 147 mJ/cm 2 of ultraviolet light exposure. A post-exposure bake at 65° C. for 1 min and 95° C. for 5 min was performed followed by a standard SU-8 developing process.
  • SU-8 3035 MicroChem, USA
  • the glass slide featuring the microstructures was formed and then analyzed under the microscope using a DS-Qi1Mc camera equipped with a Peltier cooling device and a programmable gain amplifier (Nikon) coupled to an inverted microscope equipped with a digital control module.
  • the software used was NIS-Elements Basic Research software (Br, version 4.20.00, 64 bit, Nikon).
  • a cell was considered to be CTC if it (1) expressed CK (an internal architectural protein that is largely associated with epithelial cells), (2) was characterized by a high nuclear: cytoplasmic ratio (upon DAPI staining), and (3) did not express CD45.
  • CTCs The role of CTCs was investigated in response to treatment effectiveness assessment by analyzing blood samples (1 mL) before and after chemotherapy in five patients with advanced or metastatic CCA for a period of six months.
  • the presence and number of CTCs in blood were assessed using the prior mentioned MB-SCH45 alongside MB-anti-EpCAM followed by immunofluorescence staining along with the conventional FACS and tumor size measurements were performed using CT scan.
  • CT scan measurements the solid tumors were measured in at least one dimension with the longest diameter as per RECIST guideline.
  • baseline studies of CTC enumeration from blood and radiographic analysis were performed within an average of 3 days prior to starting treatment. Blood samples from five CCA patients were obtained at 4-12-week intervals and the same type of imaging studies were performed with an average of 4 days between imaging and the blood draw.
  • microfluidic chip In order to efficiently capture cancerous cells by using magnetic beads, an integrated microfluidic chip was constructed and used in the present study.
  • the microfluidic chip was composed of 4 polydimethylsiloxane (PDMS) layers and a double-sided tape layer ( FIG. 1 ).
  • the uppermost PDMS of the chip was used for collection of cell pellet from blood, and the second and third layers were the air control and the liquid layer, respectively.
  • the fourth (bottom) layer of the chip was the thinnest and provided enough support to keep the contents of the liquid channel intact.
  • the chip was capable of performing various fluidic functions (e.g., mixing, transportation and etc.) using two types of air, compressed air and suction pressure (i.e., vacuum).
  • the flow of compressed air or generation of suction pressure was controlled by electromagnetic valves (EMVs).
  • EMVs electromagnetic valves
  • the entire set up for operation of the chip was consisted of an air compressor, a vacuum pump, and a computer controlled EMV functional unit.
  • the microfluidic chip as depicted in FIG. 1A featured two closed type micromixer/micropump/transportation units.
  • blood after lysis was dispensed into the topmost PDMS layer through the inlet (i) of the chip.
  • the channel labelled (iii) served as vent for any bubbles trapped during dispensing lysed blood into the inlets, it also served as waste collection unit for the lysed blood after collection of the pellet.
  • the chamber C 1 was designed such as to ensure no pellet loss after collection and direct transfer of the afore-said to the micromixer I of the liquid channel layer (L 2 ).
  • the second and third PDMS layer (L 2 and L 3 ) were used for positive enrichment and immunostaining procedures operated pneumatically via active mechanical displacement of two microvalves and an actuation chamber.
  • the air control layer of the chip was divided into two parts: the first and second micromixers.
  • the first micromixer (I in FIG. 1A ) served as the blood treatment unit, in which negative depletion (using magnetic beads coated with anti-CD45 (MB-CD45) for removal of white blood cells (WBCs)) and positive enrichment using magnetic beads coated with the present compound (MB-SCH45) or the anti-EpCAM (MB-anti-EpCAM) were carried out.
  • the second micromixer (II in FIG. 1A ) served as the immunofluorescence staining chamber.
  • the microfluidic chip constructed above was employed to capture cholangio-cancerous cells from healthy or CCA positive patient blood. Briefly, the blood samples collected from 65 CCA patients was independently mixed with red blood cell (RBC) lysis buffer and dispensed into 5 inlets in the topmost PDMS layer of the chip (L 1 in FIG. 1A ), then magnetic beads coated with the compound of formula (I) or anti-EpCAM were manually pipetted into the blood treatment unit, and gently mixed for 15 min. After incubation, the cells were identified and bound by the magnetic beads pre-coated with the compound of formula (I) of the present invention. Then, these magnetic beads-cells complexes were collected with a magnetic force and washed by 1 ⁇ PBS buffer, and transferred to immunofluorescence staining unit of the chip for further analysis.
  • RBC red blood cell
  • the present compounds were respectively synthesized in accordance with steps as described in schemes 1 to 5.
  • tetrasaccharide backbones were assembled in a [2+2] fashion from the appropriate disaccharide building blocks and convergent [4+4] glycosylation provided the fully protected octasaccharide skeleton, which was further subjected to functional group transformations to yield final compound with varying levels of O-sulfonation states.
  • the glycan molecule was then biotinylated such that the resulting compound 20 (i.e., SCH45), could be conjugated onto the surface of streptavidin-coated Dynabeads.
  • the structure of the present synthetic glycosaminoglycan is composed of four variably sulfated disaccharide units, with each sugar in the disaccharide unit being any one of an N-acetyl- ⁇ -D-glucosamine ( ⁇ -D-G1cMAc, abbreviated as “N”), a ⁇ -D-glucuronic acid ( ⁇ -D-G1cA, abbreviated as “G”) or an ⁇ -L-iduronic acid ( ⁇ -D-IdoA, abbreviated as “I”), and linked with another sugar by ⁇ -1 ⁇ 4 linkage.
  • N N-acetyl- ⁇ -D-glucosamine
  • G1cA ⁇ -D-G1cA
  • I ⁇ -L-iduronic acid
  • Trimethyl(4-methylphenylthio)silane (TMSSTol, 0.08 mL, 0.39 mmol) and ZnI 2 (0.13 g, 0.41 mmol) were added to a solution of compound 9 (0.43 g, 0.196 mmol) in anhydrous CH 2 C 12 (5 mL) at ambient temperature under argon. After the tetrasaccharide was consumed completely (2 h), the mixture was diluted with CH 2 C 12 (8 mL) and then filtered out. The filtrate was washed with satd. NaHCO 3(aq) , dried over MgSO 4 and filtered.
  • the reaction was warmed up to room temperature gradually and kept stirring until the complete consumption of the thioglycoside by TLC plate analysis (about 2 h).
  • the mixture was neutralized by addition of Et 3 N, diluted with CH 2 Cl 2 and filtered through Celite.
  • the resulting solution was washed with 10% Na 2 S 2 O 3(aq) and H 2 O.
  • the organic layer was combined, dried over MgSO 4 and concentrated under reduced pressure. The residue was purified by flash column chromatography to yield octasaccharides 13 (72%).
  • TEMPO 2,2,6,6-tetramethyl-1-piperidinyloxyl free radical
  • Example 2 CTC Detection by Use of Magnetic Beads Pre-Coated with the Compound of Example 1
  • a microfluid chip for capturing CTCs from 65 advanced or metastatic CCA samples was constructed, and CTCs in all 65 bold samples were respectively captured using magnetic beads pre-coated with the compound of Example 1 (i.e., MB-SCH45) or an EpCAM antibody (i.e., MB-anti-EpCAM, as a control) in accordance with the procedures described in the “Materials and Methods” section.
  • MB-SCH45 was capable of isolating CTCs from all samples, regardless of the disease status.
  • the number of CTCs isolated from the above samples using MB-SCH45 and MB-anti-EpCAM and details regarding the stage and organ of metastasis have been presented in Table 1. It is noteworthy that CTCs could be detected even in the blood of patients (Nos. 5, 14, 23, 30, 32, 43, and 60 in Table 1) with no distant metastasis as observed by radiographic imaging at the time of observation.
  • the size of the CTCs ranged from 5 to 20 ⁇ m in diameter (data not shown). Most of the cells presented a high nucleus-to-cytoplasmic ratio but not in all cases.
  • FIG. 2 The immunofluorescence images of some samples are shown in FIG. 2 . It is worth mentioning that the samples were randomly selected and in no particular order. CTC clusters could be observed using both MB-SCH4 and MB-anti-EpCAM in some stage IV patients. Some clusters were relatively small while others comprised dozens of tumor cells. The CTC clusters were more evident when MB-anti-EpCAM was used for positive enrichment and have been shown in FIG. 2 (the bottom-most 4 panels). Such cluster-forming CTCs are known to have a relatively higher potential for metastasis and should be studied in detail in future works.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Materials Engineering (AREA)
  • Inorganic Chemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Organic Chemistry (AREA)
  • Polymers & Plastics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed herein is a novel compound of formula (I) for detecting circulating cancerous cells, particularly, cholangio-cancerous cells, from a biological sample,wherein, R1 and R2 are independently H, or —SO3M; and M is a monovalent cation selected from the group consisting of sodium ion, potassium ion, lithium ion or ammonium ion. Also disclosed herein is a method of treating and detecting cholangio-cancerous cells from a biological sample of a subject suspected of having cholangiocarcinoma (CCA). The method includes steps of, contacting the biological sample with a magnetic bead pre-coated with the compound of formula (I); detecting a complex formed between the magnetic bead and the biological sample in an immunoassay; and administering to the subject an effective amount of a chemotherapeutic agent to ameliorate symptoms associated with the CCA.

Description

    STATEMENT REGARDING PRIOR DISCLOSURES BY THE INVENTOR OR A JOINT INVENTOR UNDER 37 C.F.R. 1.77(B)(6)
  • Part of the subject matter of the invention described in the present application was published by the inventors, Shang-Cheng Hung, Gwo-Bin Lee, and Priya Gopinathan in an article titled “Exploring Circulating Tumor Cells in Cholangiocarcinoma Using a Novel Glycosaminoglycan Probe on a Microfluidic Platform.” The article was published in Advanced Healthcare Materials 2020, 9: 1901875. The publication was made by and/or originated from 3 members of the inventive entity of the present invention, and the entirety of this article is incorporated herein by reference. A copy of the article is provided in a concurrently filed Information Disclosure Statement pursuant to the guidance of 78 Fed. Reg. 11076 (Feb. 14, 2013).”
  • BACKGROUND OF THE INVENTION 1. Field of the Invention
  • The present disclosure in general relates to compounds and methods for the detection of circulating cancerous cells in a biological sample.
  • 2. Description of Related Art
  • Cholangiocarcinoma (CCA) is the second most common primary malignant liver cancer occurring at the bile ducts which drain bile juice from the liver into the small intestine. It is typically characterized by late-stage diagnosis, poor prognosis, high recurrence and frequent metastasis. Nowadays, imaging techniques, including ultrasonography, magnetic resonance imaging and computed tomography, and pathological diagnoses by forceps biopsy or brush cytology are the main tools applied to detect CCA. These methods are limited by tumor sizes, localization and lesions so that they are unable to discover CCA tumor at the early stage. The most common tumor biomarkers used to diagnose CCA are carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA). However, the sensitivity and specificity of CA19-9 and CEA are still not satisfactory in clinical applications.
  • Circulating tumor cells (CTCs) have been suggested to play a role in the metastatic spread of carcinomas, accordingly, they may serve as markers for the detection of tumors, particularly, those that metastasized. However, their application in the clinical assessment of CCA remains inclusive. Quantification of a relatively low number of CTCs against a background of millions of blood cells has thwarted progress in CCA diagnosis, and affinity-based isolation techniques using CTC-specific antigens such as EpCAM, EphB4, HER2, EGFR, CEA and etc failed to produce results suffice for clinical applications.
  • Therefore, three exists in the related art a need of new biomarkers for detection CTCs in a biological sample, which in turn may provide prognosis on CCA.
  • SUMMARY OF THE INVENTION
  • Inventors of the present invention unexpectedly identify novel octasaccharides that bind to the surface of cholangio-cancerous cells, thus these octasaccharides are useful for detecting cholangio-cancerous cells in a biological sample of a subject, whom is at risk of developing cholangiocarcinoma (CCA) or has CCA.
  • Accordingly, the first aspect of the present disclosure is directed to a compound of formula (I),
  • Figure US20220373552A1-20221124-C00003
  • Wherein R1 and R2 are independently H, or —SO3M; and M is a monovalent cation selected from the group consisting of sodium ion, potassium ion, lithium ion or ammonium ion.
  • According to one preferred embodiment of the present disclosure, in the formula (I), R1 and R2 are independently —SO3M, and M is the sodium ion.
  • The second aspect of the present disclosure is directed to a method of treating and detecting circulating cholangio-cancerous cells from a biological sample of a subject suspected of having CCA. The method includes the steps of,
      • (a) contacting the biological sample with a compound of formula (I); and
      • (b) detecting a complex formed between the compound of formula (I) and the biological sample in an immunoassay; and
      • (c) administering to the subject an effective amount of a chemotherapeutic agent to ameliorate symptoms associated with the CCA.
  • According to preferred embodiments of the present disclosure, the compound of formula (I) is coupled to streptavidin, which is conjugated to the outer surface of a magnetic bead.
  • According to preferred embodiments of the present disclosure, in the compound of formula (I), R1 and R2 are independently —SO3M, and M is the sodium ion.
  • Examples of the biological sample suitable for use in the present method include, but are not limited to, blood, plasma, serum, urine, sputum, saliva, tissue sample, biopsy, and tissue lysate. Preferably, the biological sample is the blood from the subject.
  • According to preferred embodiments of the present disclosure, the subject has stage III or IV CCA, or metastatic CCA.
  • According to optional embodiment of the present disclosure, the method further comprises, before the step (a), pre-treating the blood with a lysis buffer, so as to lyse red blood cells therein.
  • Examples of the chemotherapeutic agent suitable for use in the present method include, but are not limited to, S-1, leucovorin, oxaliplatin, gemcitabine, cisplatin, and a combination thereof. In some embodiments, the method comprises administering to the subject an effective amount of the S-1 to ameliorate symptoms associated with the CCA, in which the S-1 is a combination of tegafur, 5-chloro-2,4-dihydropyrimidine (CDHP), and potassium oxonate. In other embodiments, the method comprises administering to the subject an effective amount of a combination of gemcitabine and cisplatin to ameliorate symptoms associated with the CCA. In still further embodiments, the method comprises administering to the subject an effective amount of a combination of S-1, leucovorin, oxaliplatin, and gemcitabine to ameliorate symptoms associated with the CCA.
  • According to optional embodiment of the present disclosure, the method further comprises administering to the subject a radiotherapy in addition to the administration of S-1 to ameliorate symptoms associated with the CCA.
  • The details of one or more embodiments of this disclosure are set forth in the accompanying description below. Other features and advantages of the invention will be apparent from the detail descriptions, and from claims.
  • It is to be understood that both the foregoing general description and the following detailed description are by examples, and are intended to provide further explanation of the invention as claimed.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The accompanying drawings, which are incorporated in and constitute a part of the specification, illustrate various example systems, methods and other exemplified embodiments of various aspects of the invention. The present description will be better understood from the following detailed description read in light of the accompanying drawings, where,
  • FIG. 1A: Schematic diagrams of a microfluidic chip. The chip features all layers from L1 to L5 representing liquid channel layer, air control layer, liquid channel layer, thin PDMS layer and double-sided tape, respectively. Detail structure of layers L1, L2 and L3 are shown where (i) represents inlets for lysed blood, (ii) represents pellet collection area, and (iii) represents outlets for waste collection. (I) represents the blood treatment unit, and (II) represents the immunofluorescence staining unit. The 13 connections to electromagnetic valves (EMVs) for pneumatic control of the chip are shown as 1 to 13. The 8 microchambers/reagent storing units are labeled as C1 to C8. C1: cell pellet collection unit, C2: waste collection unit, C3: magnetic bead storage unit, C4 and C5: 1× phosphate-buffered saline (PBS) storage unit, C6: the storage unit for secondary antibody (i.e., DyLight 488 and CD45-PE first and later for DAPI), C7: waste collection unit, and C8: anti-CK17 primary antibody storage unit.
  • FIG. 1B: Photograph of the microfluidic chip of FIG. 1A.
  • FIG. 2: Photographs depicting immunofluorescence staining of CTC clusters captured by MB-SCH45 or MB-anti-EpCAM. Negative depletion with anti-CD45 Ab and positive enrichment with MB-SCH45 (top 16 panels) and MB-anti-EpCAM (lower 20 panels) for both representative stage III and IV samples. The bottom 4 panels show the CTC culsters after positive enrichment with MB-anti-EpCAM.
  • FIG. 3: The predictive significance of CTC counts. The correlation between CTC count and clinical response to chemotherapy in 5 advanced or metastatic CCA patients was tested. Panels (A) to (E) represent patients A to E. Subpanels I-III in each panel represent normalized CTC counts isolated using MB-SCH45, MB-anti-EpCAM and FACS, respectively. Subpanel IV represents the normalized tumor size using radiographic imaging.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The detailed description provided below in connection with the appended drawings is intended as a description of the present disclosure and is not intended to represent the only forms in which the present disclosure may be constructed or utilized.
  • 1. Definitions
  • Unless otherwise indicated, the term “patient” or “subject” may be used interchangeably in the present disclosure, and refers to any animal. The animal can be a human subject, or a non-human subject. The subject may be a human, but can also be a mammal in need of veterinary treatment, e.g., domestic animals or game animals, farm animals, and laboratory animals (e.g., rats, mice, guinea pigs, primates, and the like). Usually the animal is a non-human mammal, such as a non-human primate. Non-human primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus or Pan. Domestic animals and game animals include cows, horses, pigs, sheep, deer, bison, buffalo, mink, felines (e.g., domestic cats, canines (e.g., dogs)), wolf and fox, avian species (e.g., chicken, turkey, and ostrich), and fish (e.g., trout, catfish, and salmon).
  • The term “contacting” is used herein with respect to a cell (e.g., a cell in a biological sample) and refers to any mode of delivery or “administration” of an agent either to cells or the biological sample, in which the agent (e.g., a compound of the present disclosure or a magnet bead pre-coated with the compound of the present disclosure) is brought into contact with one or more cells in sufficient amount to achieve affinity binding therebetween.
  • Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in the respective testing measurements. Also, as used herein, the term “about” generally means within 10%, 5%, 1%, or 0.5% of a given value or range. Alternatively, the term “about” means within an acceptable standard error of the mean when considered by one of ordinary skill in the art. Other than in the operating/working examples, or unless otherwise expressly specified, all of the numerical ranges, amounts, values and percentages such as those for quantities of materials, durations of times, temperatures, operating conditions, ratios of amounts, and the likes thereof disclosed herein should be understood as modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the present disclosure and attached claims are approximations that can vary as desired. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
  • The singular forms “a”, “and”, and “the” are used herein to include plural referents unless the context clearly dictates otherwise.
  • 2. The Compound of the Present Invention
  • Aspects of the present disclosure relate to the finding that a novel octasaccharide is useful as a biomarker for the identification and/or detection of cancerous cells in a biological sample. The novel octasaccharide is described herein.
  • In one aspect, the present invention relates to a compound of formula (I):
  • Figure US20220373552A1-20221124-C00004
  • Wherein, R1 and R2 are independently H, or —SO3M; and M is a monovalent cation selected from the group consisting of sodium ion, potassium ion, lithium ion or ammonium ion.
  • In some embodiments of the present disclosure, in the formula (I), R1 and R2 are independently —SO3M, and M is the sodium ion.
  • The compound of the present disclosure may be prepared in accordance with procedures described in the working examples. All stereoisomers of the present compounds, such as those which may exist due to asymmetric carbons on the R substituents of the compound of formula (I) including enantiomeric and diastereomeric forms, are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • 3. Method of Use
  • The compound of formula (I) may bind to surfaces of cancerous cells, particularly cancers originated from cholangio bile duct. Accordingly, the present disclosure thus encompasses a method of identifying or detecting cholangio-cancerous cells from a biological sample of a subject suspected of having cholangiocarcinoma (CCA), as well as a method of treating the subject based on the detection of cholangio-cancerous cells in the biological sample.
  • The present method includes contacting a biological sample of the subject with a compound of formula (I), in which a binding between the compound of formula (I) and the biological sample indicates the presence of cholangio-cancerous cells in the biological sample. Alternatively, or optionally, for better identification or detection of cholangio-cancerous cells in the blood sample, the blood sample is pre-treated with a lysis buffer, which will result in the lysis of red blood cells in the blood sample, prior to commencing the present method (i.e., prior to contacting the blood with magnetic beads). Examples of the biological sample suitable for use in the present method, include but are not limited to, blood, plasma, serum, urine, sputum, saliva, tissue sample, biopsy, and tissue lysate. In one preferred embodiment, the biological sample is blood. Accordingly, the method comprises mixing a blood sample of the subject with the present compound. Upon mixing, if the blood sample does contain cholangio-cancer cells, they will automatically bind with the compound of formula (I) and form a complex, which may be detected via any suitable means, such as enzyme-linked immunosorbent assay (ELISA), immunofluorescent staining and the like; and the presence of the complex is an indication that the subject is positive for CCA. By contrast, if the blood sample does not contain cholangio-cancer cells, there will not be a binding between the blood sample and the compound of formula (I), and the absence of the binding is an indication that the subject is negative for CCA. Preferably, the compound of formula (I) is coated on the surface of a magnetic bead via coupling to streptavidin pre-conjugated on the outer surface of the magnetic bead. Thus, upon mixing with the biological sample, the magnetic beads having the compound of formula (I) coated thereon would be able to capture the cholangio-cancerous cells present in the biological sample, and the magnetic beads having cholangio-cancerous cells captured thereon may be allocated and harvested via use of a magnet.
  • Upon confirming the subject's disease status, particularly, when the subject is positive for CCA, suitable treatment (e.g., radiotherapy, chemotherapy and the like) may then be administered to the subject to ameliorate symptoms associated with the CCA. In some embodiments, the method therefore includes administering an effective amount of a chemotherapeutic agent to the subject. Examples of the chemotherapeutic agent suitable for use in the present method include, but are not limited to, S-1, leucovorin, oxaliplatin, gemcitabine, cisplatin, and a combination thereof. In some embodiments, an effective amount of the S-1 is administered to the subject to ameliorate symptoms associated with the CCA, in which the S-1 is a combination of tegafur, 5-chloro-2,4-dihydropyrimidine (CDHP), and potassium oxonate. In optional embodiments, a radiotherapy is administered to the subject before, concurrently with, or after the administration of the S-1. In other embodiments, a combination of gemcitabine and cisplatin is administered to the subject to ameliorate symptoms associated with the CCA. In further embodiments, a combination of S-1, leucovorin, oxaliplatin, and gemcitabine is administered to the subject to ameliorate symptoms associated with the CCA.
  • According to preferred embodiments of the present disclosure, the subject has stage III or IV CCA, or metastatic CCA.
  • The present invention will now be described more specifically with reference to the following embodiments, which are provided for the purpose of demonstration rather than limitation. While they are typically of those that might be used, other procedures, methodologies, or techniques known to those skilled in the art may alternatively be used.
  • EXAMPLES
  • Conjugation of the Present Compound to Magnetic Beads
  • Compound 20 (100×10−6 M) was incubated with Dynabeads MyOne streptavidin C1 (≈7−10×109 beads mL−1, Ø=1 μm, Invitrogen, USA) magnetic beads in a volume/volume ratio of 1:10 and placed on an RM-2L INTELLI-mixer (ELMI Ltd., Latvia) at 25 rpm for 30 min at room temperature (C2 mode). The beads were then collected using a magnetic particle concentrator (MPC, Dynabeads MPC-1, Life Technologies) for 2 min. The supernatant was discarded, and the beads were washed thrice with 1 mL of deionized water (DI water). The bead conjugates MB-SCH45 were suspended in the same volume of DI water as the initial volume of Dynabeads used.
  • Blood Specimen Collection
  • Informed written consent was obtained from all 65 advanced stage or metastatic CCA patients. The three healthy blood samples used in this study were collected from Tainan blood bank, Taiwan. For each donor, peripheral blood (10 mL) was drawn into BD Vacutainer spray coated K2EDTA tubes (BD Diagnostics, USA). All samples were maintained and transported at 4° C. and processed within 30 h of collection. To avoid bias, samples were blindly processed without prior information on the disease status, cancer stage, or metastasis in patients.
  • Experimental Set-Up
  • The experimental procedure involved four steps: (1) RBC lysis and isolation of pellet, (2) negative depletion for the removal of WBCs, (3) positive magnetic enrichment, and 4) immunofluorescence staining.
  • (1) RBC Lysis and Isolation of Pellet
  • RBC were lysed by adding 9 mL of 1× RBC lysis buffer (Biolegend, USA) to 1 mL of peripheral blood and incubating at room temperature with gentle mixing on a rotary shaker (TKS ROCKER-S-01, Lab666, Taiwan) for 5 min. Then the sample was dispensed into the topmost PDMS layer of the chip through the inlets provided (FIG. 1A). The microfluidic chip was then spun as described above and placed on a PMMA plate with a groove such that the microfluidic chip could be placed inside it. It was then placed on a spin-coater (M and R Nanotechnology Co. Taoyuan, Taiwan) and spun at 1,200 rpm for 2 min at a negative gauge pressure of −67 kPa and the cell pellet was then moved into the blood treatment unit of the chip's liquid channel layer by a vacuum force created by the microvalves. The waste blood with lysed RBCs was removed through the waste outlets (marked as (iii) in FIG. 1A) using vacuum suction. The remaining 5 mL of lysed blood was then dispensed into the topmost layer of the microfluidic chip and the above process was repeated. The pellet was transported to the micromixer and suspended in 180 μL of 1× phosphate buffered saline (PBS) by activating the micromixer for 2 min. The resulting cell suspension consisting of predominately nucleated cells was then subjected to negative depletion for the removal of leucocytes.
  • (2) Negative Depletion and Positive Magnetic Enrichment
  • A negative depletion strategy reliant on the removal of leucocytes via their binding to anti-CD45 Ab bound to magnetic beads (Dynabeads CD45, Thermo-Fisher Scientific) was employed herein on-chip. The cell-bead complexes were then attracted to a magnet to remove the bound leukocytes from the lysed blood samples. After four rounds of negative depletion, the remaining cells (predominantly CTCs) in the collected supernatant were mixed with MB-SCH45 (bead conjugated with compound 20 described above) or MB-anti-EpCAM (EpiEnrich, 4×108 beads mL−1, 0=4.5 μm, Thermo-Fisher Scientific). The magnetic bead-CTC complexes were then collected, washed, and transferred to the immunofluorescence staining unit of the chip for further analysis.
  • (3) Immunofluorescence Staining
  • For immunofluorescence staining, primary anti-CK17 (1:500 dilution; 100 μL, 0.6 mg mL−1, GTX103765, Rabbit, GeneTex, USA) was used. The secondary Ab, goat antirabbit IgG DyLight 488; GeneTex and anti-CD45-PE (RRID: AB_10375163, HUCD45 PE, Life Technologies) were used at a dilution of 1:500. It is to be noted here that the anti-CD45 Ab used had a PE tag thereby requiring no primary Ab. DAPI (1 mg mL−1, Thermo Fischer, US) was then used as the nuclear stain. After the final magnetic collection, the bead-cell complexes were suspended in 10 μL of 1×PBS and collected through the waste collection chamber. They were transferred onto the numbered microscopic slide and ProLong Gold Antifade Reagent (Invitrogen) was used to embed the cell-bead complexes.
  • The numbered microscope slide was fabricated using an SU-8 standard photolithography process to facilitate CTC counting. Concisely, a 30 μm thick layer of SU-8 3035 (MicroChem, USA) was spin-coated on a glass microscope slide. A soft-baking process for 10 min was performed and the SU-8 microstructures were patterned at a dosage of 147 mJ/cm2 of ultraviolet light exposure. A post-exposure bake at 65° C. for 1 min and 95° C. for 5 min was performed followed by a standard SU-8 developing process. The glass slide featuring the microstructures was formed and then analyzed under the microscope using a DS-Qi1Mc camera equipped with a Peltier cooling device and a programmable gain amplifier (Nikon) coupled to an inverted microscope equipped with a digital control module. The software used was NIS-Elements Basic Research software (Br, version 4.20.00, 64 bit, Nikon). A cell was considered to be CTC if it (1) expressed CK (an internal architectural protein that is largely associated with epithelial cells), (2) was characterized by a high nuclear: cytoplasmic ratio (upon DAPI staining), and (3) did not express CD45.
  • Assessment of CTCs in Metastatic CCA Patients before and after Chemotherapy
  • The role of CTCs was investigated in response to treatment effectiveness assessment by analyzing blood samples (1 mL) before and after chemotherapy in five patients with advanced or metastatic CCA for a period of six months. The presence and number of CTCs in blood were assessed using the prior mentioned MB-SCH45 alongside MB-anti-EpCAM followed by immunofluorescence staining along with the conventional FACS and tumor size measurements were performed using CT scan. For CT scan measurements, the solid tumors were measured in at least one dimension with the longest diameter as per RECIST guideline. Before initiating chemotherapy, baseline studies of CTC enumeration from blood and radiographic analysis were performed within an average of 3 days prior to starting treatment. Blood samples from five CCA patients were obtained at 4-12-week intervals and the same type of imaging studies were performed with an average of 4 days between imaging and the blood draw.
  • Construction of a Microfluidic Chip
  • In order to efficiently capture cancerous cells by using magnetic beads, an integrated microfluidic chip was constructed and used in the present study. The microfluidic chip was composed of 4 polydimethylsiloxane (PDMS) layers and a double-sided tape layer (FIG. 1). The uppermost PDMS of the chip was used for collection of cell pellet from blood, and the second and third layers were the air control and the liquid layer, respectively. The fourth (bottom) layer of the chip was the thinnest and provided enough support to keep the contents of the liquid channel intact. All layers were bonded together via oxygen plasma treatment (FC-12064, FEMTO Science, USA) at 90 W for 1 min, which also introduced polar functional silanol (SiOH) groups, changing the surface properties of PDMS from being hydrophobic to hydrophilic. The chip was then treated with a solution of Pluronic P123 (a triblock copolymer of poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide)) in 99% (by weight) aqueous ethanol to increase hydrophilicity of PDMS. P123 was dispensed into the topmost layer of the chip and allowed to react with PDMS for 5 min. Microvalves were actuated so that P123 solution was transported to the chip's liquid layer and finally through the waste collection chambers. The chip was capable of performing various fluidic functions (e.g., mixing, transportation and etc.) using two types of air, compressed air and suction pressure (i.e., vacuum). The flow of compressed air or generation of suction pressure was controlled by electromagnetic valves (EMVs). The entire set up for operation of the chip was consisted of an air compressor, a vacuum pump, and a computer controlled EMV functional unit.
  • The microfluidic chip as depicted in FIG. 1A featured two closed type micromixer/micropump/transportation units. As shown in FIG. 1A, blood after lysis was dispensed into the topmost PDMS layer through the inlet (i) of the chip. The channel labelled (iii) served as vent for any bubbles trapped during dispensing lysed blood into the inlets, it also served as waste collection unit for the lysed blood after collection of the pellet. The chamber C1 was designed such as to ensure no pellet loss after collection and direct transfer of the afore-said to the micromixer I of the liquid channel layer (L2). The second and third PDMS layer (L2 and L3) were used for positive enrichment and immunostaining procedures operated pneumatically via active mechanical displacement of two microvalves and an actuation chamber. The air control layer of the chip was divided into two parts: the first and second micromixers. The first micromixer (I in FIG. 1A) served as the blood treatment unit, in which negative depletion (using magnetic beads coated with anti-CD45 (MB-CD45) for removal of white blood cells (WBCs)) and positive enrichment using magnetic beads coated with the present compound (MB-SCH45) or the anti-EpCAM (MB-anti-EpCAM) were carried out. The second micromixer (II in FIG. 1A) served as the immunofluorescence staining chamber.
  • Capturing Cholangio-Cancerous Cells Using the Microfluidic Chip
  • The microfluidic chip constructed above was employed to capture cholangio-cancerous cells from healthy or CCA positive patient blood. Briefly, the blood samples collected from 65 CCA patients was independently mixed with red blood cell (RBC) lysis buffer and dispensed into 5 inlets in the topmost PDMS layer of the chip (L1 in FIG. 1A), then magnetic beads coated with the compound of formula (I) or anti-EpCAM were manually pipetted into the blood treatment unit, and gently mixed for 15 min. After incubation, the cells were identified and bound by the magnetic beads pre-coated with the compound of formula (I) of the present invention. Then, these magnetic beads-cells complexes were collected with a magnetic force and washed by 1× PBS buffer, and transferred to immunofluorescence staining unit of the chip for further analysis.
  • Statistical Analysis: The sample size (n) has been presented in the pertinent figure legends. Details regarding data normalization (for before and after chemotherapy patients) have been described in the Experimental Section. Statistical analysis was performed using OriginPro (OriginLab, Massachusetts, USA).
  • Example 1 Chemical Synthesis of the Present Compound
  • The present compounds were respectively synthesized in accordance with steps as described in schemes 1 to 5. In general, tetrasaccharide backbones were assembled in a [2+2] fashion from the appropriate disaccharide building blocks and convergent [4+4] glycosylation provided the fully protected octasaccharide skeleton, which was further subjected to functional group transformations to yield final compound with varying levels of O-sulfonation states. The glycan molecule was then biotinylated such that the resulting compound 20 (i.e., SCH45), could be conjugated onto the surface of streptavidin-coated Dynabeads.
  • The structure of the present synthetic glycosaminoglycan (GAC) is composed of four variably sulfated disaccharide units, with each sugar in the disaccharide unit being any one of an N-acetyl-α-D-glucosamine (α-D-G1cMAc, abbreviated as “N”), a β-D-glucuronic acid (β-D-G1cA, abbreviated as “G”) or an α-L-iduronic acid (α-D-IdoA, abbreviated as “I”), and linked with another sugar by α-1→4 linkage. Thus, the present compound 20 may be represented as NINGNING.
  • Figure US20220373552A1-20221124-C00005
  • Figure US20220373552A1-20221124-C00006
  • Figure US20220373552A1-20221124-C00007
  • Figure US20220373552A1-20221124-C00008
  • Figure US20220373552A1-20221124-C00009
  • Figure US20220373552A1-20221124-C00010
    Figure US20220373552A1-20221124-C00011
    Figure US20220373552A1-20221124-C00012
    Figure US20220373552A1-20221124-C00013
    Figure US20220373552A1-20221124-C00014
  • 6-O-Acetyl-4-O-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-4-O-(2-naphthylmethyl)-α-D-glucopyranosyl]-2-O-benzoyl-3-O-benzyl-β-D-glucopyranosyl acetate (7)
  • A solution of the thioglycoside 1 (193 mg, 0.22 mmol) and the 4-alcohol 6 (86 mg, 0.19 mmol) in anhydrous CH2Cl2 (5.6 mL) with freshly dried 3 Å molecular sieves (557 mg) was stirred at room temperature for 1 h under argon. The reaction flask was cooled down to −60° C., N-idosuccinimide (NIS, 61 mg, 0.26 mmol) and triflic acid (TfOH, 3 μL, 0.04 mmol) were added to the solution, and the mixture was gradually warmed up to −50° C. After stirring for 2 h, the reaction was quenched by addition of Et3N (8 μL, 0.06 mmol) was added to quench the reaction, the mixture was filtered through Celite, and the solid was washed by CH2Cl2. The filtrate was washed with 10% Na2S2O5(aq) (5.0 mL), dried over MgSO4, filtered and concentrated under reduced pressure to furnish a residue, which was purified by flash column chromatography (ethyl acetate/hexane=1/4) to give the expected disaccharide 34 (192 mg, 86%). [α]25 D+0.6 (c 5.34, CHCl3); IR (thin film): v 2930, 2107, 1738, 1265, 1111, 1067 cm−1; 1H NMR (600 MHz, CDCl3): δ 8.02 (d, J=7.6 Hz, 2H, Bz-H), 7.85-7.81 (m, 1H, Ar—H), 7.79-7.75 (m, 2H, Ar—H), 7.66 (d, J=7.1 Hz, 2H, Ar—H), 7.64-7.61 (m, 3H, Ar—H), 7.58 (t, J=7.4 Hz, 1H, Ar—H), 7.51-7.47 (m, 2H, Ar—H), 7.45 (t, J=7.8 Hz, 2H, Ar—H), 7.42 (d, J=8.2 Hz, 2H, Ar—H), 7.40-7.34 (m, 2H, Ar—H), 7.34-7.30 (m, 2H, Ar—H), 7.30-7.24 (m, 5H, Ar—H), 7.24-7.20 (m, 3H, Ar—H), 7.18 (d, J=8.2 Hz, 2H, Ar—H), 5.83 (d, J=7.9 Hz, 1H, 1-H), 5.52 (d, J=3.6 Hz, 1H, 1′-H), 5.43 (dd, J=7.9, 8.5 Hz, 1H, 2-H), 4.95-4.88 (m, 2H, ArCH2), 4.85-4.80 (m, 2H, ArCH2), 4.76 (d, J=10.7 Hz, 1H, ArCH2), 4.73 (d, J=10.7 Hz, 1H, ArCH2), 4.35 (d, J=12.3 Hz, 1H, 6-Ha), 4.09 (dd, J=12.3, 4.1 Hz, 1H, 6-Hb), 4.04 (dd, J=8.5, 8.5 Hz, 1H, 3-H), 4.02-3.80 (m, 1H, 6′-Ha), 3.95 (dd, J=9.0, 8.5 Hz, 1H, 4-H), 3.92-3.88 (m, 2H, 3′-H, 5′-H), 3.81-3.77 (m, 1H, 5-H), 3.75 (d, J=11.7 Hz, 1H, 6′-Hb), 3.62 (d, J=6.8 Hz, 1H, 4′-H), 3.32-3.25 (m, 1H, 2′-H), 1.99 (s, 3H, Ac), 1.74 (s, 3H, Ac), 1.04 (s, 9H, tBu TBDPS); 13C NMR (150 MHz, CDCl3): δ 170.4 (C), 169.2 (C), 165.0 (C), 137.3 (C), 136.7 (C), 135.9 (CH), 135.5 (CH), 135.3 (C), 133.6 (CH), 133.3 (C), 133.2 (C), 133.0 (C), 132.8 (C), 131.5 (CH), 129.8 (CH), 129.7 (CH), 129.5 (CH), 129.1 (C), 128.6 (CH), 128.4 (CH), 128.2 (CH), 127.9 (CH), 127.8 (CH), 127.75 (CH), 127.70 (CH), 127.6 (CH), 126.3 (CH), 126.2 (CH), 126.0 (CH), 125.5 (CH), 121.8(C), 97.9 (CH), 91.7 (CH), 82.9 (CH), 79.9 (CH), 78.0 (CH), 75.2 (CH2), 74.6 (CH2), 74.5 (CH2), 73.4 (CH), 73.2 (CH), 73.0 (CH), 72.6 (CH), 63.3 (CH), 62.6 (CH2), 62.0 (CH2), 26.9 (CH3), 20.8 (CH3), 20.4 (CH3), 19.3 (C); HRMS ESI: m/z calcd for C64H66N3O13BrSiNa ([M+Na]+): 1214.3446, found: 1214.3455.
  • [2-Azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-4-O-(2-naphthylmethyl)-α-D-glucopyranosyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzy 1-α-L-idopyranosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsil yl-2-deoxy-α-D-glucopyranosyl]-(1→4)-6-O-acetyl-2-O-benzoyl-3-O-benzyl-β-D-gl ucopyranosyl acetate (9)
  • A solution of the thioglycoside 5 (1.2 equiv.) and the 4′-alcohol 8 (1.0 equiv.) in anhydrous CH2Cl2 (10 mL/total grams of the thioglycoside and the 4′-alcohol) with freshly activated AW-300 molecular sieves (0.5 g/total grams of the thioglycoside and the 4′-alcohol) was stirred at room temperature for 1 h and then cooled to −78° C. NIS (2.0 equiv.) was added to this mixture, and after 5 min, TfOH (0.1 equiv.) was added slowly through a microsyringe. The temperature was gradually raised to −50° C. and kept at this temperature until the complete consumption of the thioglycoside 5 by TLC plate analysis (about 2 h). Then the mixture was neutralized by addition of Et3N, diluted with CH2C12 and filtered through Celite. The resulting solution was washed with a 10% Na2S2O3(aq) and H2O. The organic layer was combined, dried over MgSO4 and concentrated under reduced pressure. The residue was purified by flash column chromatography to yield the tetrasaccharides 9 (81%).
  • IR (thin film): v 2930, 2108, 1742, 1266, 1111 cm−1; 1H NMR (600 MHz, CDCl3): δ 8.03 (dd, J=8.2, 1.0 Hz, 2H, Bz-H), 8.00 (dd, J=7.8, 1.1 Hz, 2H, Bz-H), 7.84 (dd, J=6.1, 3.1 Hz, 1H 2-NAP-H), 7.78-7.72 (m, 2H, Ar—H), 7.69-7.61 (m, 4H, Ar—H), 7.60-7.54 (m, 6H, Ar—H), 7.51-7.43 (m, 4H, Ar—H), 7.41-7.29 (m, 13H, Ar—H), 7.28-7.13 (m, 17H, Ar—H), 7.05 (d, J=8.3 Hz, 1H, Ar—H), 7.00 (d, J=8.3 Hz, 1H, Ar—H), 5.83 (d, J=8.0 Hz, 1H, 1-H), 5.44-5.39 (m, 3H, 1′-H, 1″-H, 2-H), 5.17 (s, 1H, 2″-H), 4.90 (d, J=11.3 Hz, 1H, ArCH2), 4.89 (d, J=10.8 Hz, 1H, ArCH2), 4.84 (s, 2H, ArCH2), 4.78 (d, J=10.3 Hz, 1H, ArCH2), 4.73-4.70 (m, 2H, ArCH2), 4.69 (d, J=3.6 Hz, 1H, 1′″-H), 4.48 (d, J=11.3 Hz, 1H, ArCH2), 4.39-4.33 (m, 3H, ArCH2, 6-Ha, 5″-H), 4.17 (dd, J=9.6, 8.8 Hz, 1H, 4′-H), 4.15-4.08 (m, 3H, ArCH2, 3″-H, 6″-Ha), 4.05-3.99 (m, 3H, 3-H, 6′-Ha, 6″-Hb), 3.96 (dd, J=12.1, 4.3 Hz, 1H, 6-Hb), 3.89-3.80 (m, 4H, 4-H, 6′-Hb, 4′″-H, 6″-Ha), 3.77 (ddd, J=9.3, 3.8, 2.1 Hz, 1H, 5-H), 3.74-3.70 (m, 2H, 3′-H, 5″-H), 3.68-3.63 (m, 2H, 3′″-H, 6”-Hb), 3.55 (s, 1H, 4″-H), 3.51 (d, J=9.6 Hz, 1H, 5′-H), 3.29 (dd, J=10.2, 3.5 Hz, 1H, 2′″-H), 3.18 (dd, J=10.3, 4.0 Hz, 1H, 2′-H), 1.99 (s, 3H, Ac), 1.73 (s, 3H, Ac), 1.68 (s, 3H, Ac), 1.05 (s, 9H, tBu), 0.96 (s, 9H, tBu); 13C NMR (150 MHz, CDCl3): δ 170.1 (C), 170.0 (C), 169.1 (C), 165.5 (C), 164.9 (C), 137.3 (C), 137.2 (C), 137.0 (C), 136.7 (C), 135.8 (CH), 135.57 (CH), 135.55 (CH), 135.4 (C), 133.5(CH), 133.3 (C), 133.2 (C), 133.1 (C), 133.0 (CH), 132.98 (C), 132.92 (C), 131.4 (C), 132.8(C), 131.4 (CH), 131.0 (CH), 129.8 (CH), 129.7 (CH), 129.6 (CH), 129.5 (CH), 129.4 (CH), 129.0 (C), 128.9 (CH), 128.6 (CH), 128.59 (CH), 128.52 (CH), 128.3 (CH), 128.15 (CH), 128.09 (CH), 127.85 (CH), 127.81 (CH), 127.71 (CH), 127.67 (CH), 127.57 (CH), 127.50 (CH), 127.3 (CH), 126.3 (CH), 126.1 (CH), 125.9 (CH), 125.7 (CH), 121.6 (C), 121.1 (C), 98.4 (CH), 97.4 (CH), 97.0 (CH), 91.7 (CH), 82.9 (CH), 80.3 (CH), 78.5 (CH), 77.7 (CH), 75.1 (CH2), 74.4 (CH2), 74.2 (CH2), 74.1 (CH2), 73.8 (CH), 73.4 (CH), 73.0 (CH), 72.8 (CH, CH2), 72.64 (CH), 72.59 (CH), 72.55 (CH), 68.4 (CH), 64.8 (CH), 63.9 (CH), 63.6 (CH), 62.3 (CH2), 62.2 (CH2), 62.0 (CH2), 26.8 (CH3), 26.7 (CH3), 20.8 (CH3), 20.6 (CH3), 20.3 (CH3), 19.36 (C), 19.30 (C); HRMS (MALDI): m/z calcd for C115H120Br2N6O24Si2Na ([M+Na]+): 2209.2178, found: 2209.2236.
  • 4-Methylphenyl [2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-4-O-(2-naphthyl methyl)-α-D-glucopyranosyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzyl-α-L-idop yranosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-6-O-acetyl-2-O-benzoyl-3-O-benzyl-1-thio-β-D-glucop yranoside (10)
  • Trimethyl(4-methylphenylthio)silane (TMSSTol, 0.08 mL, 0.39 mmol) and ZnI2 (0.13 g, 0.41 mmol) were added to a solution of compound 9 (0.43 g, 0.196 mmol) in anhydrous CH2C12 (5 mL) at ambient temperature under argon. After the tetrasaccharide was consumed completely (2 h), the mixture was diluted with CH2C12 (8 mL) and then filtered out. The filtrate was washed with satd. NaHCO3(aq), dried over MgSO4 and filtered. The resulting solution was concentrated under reduced pressure and purified by flash column chromatography (ethyl acetate/hexane=1/5) to provide the thioglycoside 10 (0.38 g, 85%). IR (thin film): v 2927, 2108, 1733, 1266, 1112, 708 cm−1; 1H NMR (600 MHz, CDCl3): δ 8.09 (dd, J=7.9, 1.0 Hz, 2H, Bz-H), 7.99 (d, J=8.1, 1.5 Hz, 2H, Bz-H), 7.84 (dd, J=6.0, 3.4 Hz, 1H, 2-NAP-H), 7.77-7.72 (m, 2H, Ar—H), 7.68-7.52 (m, 10H, Ar—H), 7.51-7.44 (m, 4H, Ar—H), 7.41-7.12 (m, 33H, Ar—H), 7.08-6.98 (m, 6H, Ar—H), 5.43 (d, J=4. Hz, 1H, 1′-H), 5.40 (s, 1H, 1″-H), 5.24 (dd, J=9.7, 8.7 Hz, 1H, 2-H), 5.19 (s, 1H, 2″-H), 4.90 (d, J=11.3 Hz, 1H, ArCH2), 4.89 (d, J=11.0 Hz, 1H, ArCH2), 4.84 (s, 2H, ArCH2), 4.75-4.65 (m, 5H, ArCH2, 1-H, 1′″-H), 4.49-4.44 (m, 2H, ArCH2, 6-Ha), 4.38-4.33 (m, 2H, ArCH2, 5″-H), 4.17-4.09 (m, 4H, ArCH2, 4′-H, 3″-H, 6′-Ha), 4.05-4.00 (m, 2H, 5′″-H, 6”-Ha), 3.95 (dd, J=8.3, 8.7 Hz, 1H, 3-H), 3.90 (dd, J=11.9, 4.7 Hz, 1H, 6-Hb), 3.88-3.80 (m, 3H, 6″-Hb, 4′″-H, 6′″-Ha), 3.74-3.63 (m, 5H, 4-H, 3′-H, 3′″-H, 6′-Hb, 6″-Hb), 3.60 (ddd, J=9.8, 4.8, 2.4 Hz, 1H, 5-H), 3.55 (s, 1H, 4″-H), 3.48 (d, J=9.7 Hz, 1H, 5′-H), 3.29 (dd, J=10.2, 3.5 Hz, 1H, 2′″-H), 3.15 (dd, J=10.3, 4.1 Hz, 1H, 2′-H), 2.32 (s, 3H, SPhCH3), 1.72 (s, 3H, Ac), 1.69 (s, 3H, Ac), 1.05 (s, 9H, tBu), 0.95 (s, 9H, tBu); 13C NMR (150 MHz, CDCl3): δ 170.1 (C), 170.0 (C), 165.6 (C), 165.5 (C), 165.1 (C), 138.3 (C), 137.4 (C), 137.3 (C), 137.1 (C), 136.7 (C), 135.83 (CH), 135.82 (CH), 135.57 (CH), 135.54 (CH), 135.4 (C), 133.7 (CH), 133.4 (CH), 133.31 (C), 133.27 (C), 133.2 (C), 133.1 (CH), 133.92 (C), 133.88 (C), 131.4 (C), 131.0 (CH), 129.81 (CH), 129.79 (CH), 129.70 (CH), 129.67 (CH), 129.61 (C), 129.59 (CH), 129.57 (C), 128.9 (CH), 128.6 (CH), 128.53 (CH), 128.51 (CH), 128.3 (CH), 128.16 (C), 128.12 (CH), 128.09 (CH), 127.9 (CH), 127.71 (CH), 127.67 (CH), 127.60 (CH), 127.57 (CH), 127.5 (CH), 127.4 (CH), 126.4 (CH), 126.2 (CH), 126.0 (CH), 125.7 (CH), 121.6 (C), 121.1 (C), 98.5 (CH), 97.4 (CH), 97.0 (CH), 85.9 (CH), 84.7 (CH), 80.3 (CH), 78.6 (CH), 77.7 (CH), 76.4 (CH), 75.1 (CH2), 74.6 (CH2), 74.2 (CH2), 73.9 (CH), 73.1 (CH), 72.91 (CH), 72.86 (CH, CH2), 72.7 (CH), 72.6 (CH), 68.6 (CH), 64.9 (CH), 63.9 (CH), 63.5 (CH), 62.6 (CH2), 62.3 (CH2), 62.1 (CH2), 62.0 (CH2), 26.9 (CH3), 26.8 (CH3), 21.1 (CH3), 20.6 (CH3), 20.4 (CH3), 19.4 (C), 19.3 (C); HRMS (MALDI): m/z calcd for C120H124Br2N6O22SSi2Na ([M+Na]+): 2273.3711, found: 2273.3777.
  • 4-Methylphenyl [2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyran osyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzyl-α-L-idopyrano-syl)-(1→4)-[2-azid o-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-( 1→4)-6-O-acetyl-2-O-benzoyl-3-O-benzyl-1-thio-β-D-glucopyranoside (11)
  • To a solution of the thioether-linked tetrasaccharide 10 (0.38 g, 0.17 mmol) in a mixed solvent (CH2Cl2/H2O=19/1, 11.4 mL) was added DDQ (0.06 g, 0.26 mmol) at room temperature. After stirring for 2 h, the reaction was quenched by addition of 10% Na2S2O3(aq), and the organic mixture was extracted with CH2C12. The organic layers were consecutively washed with satd. NaHCO3(aq) and H2O, dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography (ethyl acetate/hexane=1/5) to give the 4′″-alcohol 11 (0.31 g, 87%). IR (thin film): v 2929, 2108, 1733, 1265, 1112, 708 cm−1; 1H NMR (600 MHz, CDCl3): δ 8.08 (d, J=7.8 Hz, 2H, Bz-H), 8.02 (d, J=7.9 Hz, 2H, Bz-H), 7.67-7.55 (m, 9H, Ar—H), 7.49-7.39 (m, 7H, Ar—H), 7.39-7.25 (m, 17H, Ar—H), 7.23-6.15 (m, 9H, Ar—H), 7.12-7.04 (m, 6H, Ar—H), 5.44 (d, J=4.0 Hz, 1H, l′-H), 5.42 (s, 1H, 1″-H), 5.23 (dd, J=9.8, 9.1 Hz, 1H, 2-H), 5.19 (s, 1H, 2″-H), 4.90 (d, J=11.6 Hz, 1H, ArCH2), 4.84 (d, J=11.1 Hz, 1H, ArCH2), 4.75-4.65 (m, 5H, 1-H, 1″-H, ArCH2), 4.49 (d, J=11.3 Hz, 2H, ArCH2), 4.45 (d, J=10.6 Hz, 1H, 6-Ha), 4.36 (td, J=6.5, 2.2 Hz, 1H, 5″-H), 4.20 (d, J=11.1 Hz, 1H, ArCH2), 4.15 (dd, J=9.8, 9.2 Hz, 1H, 4′-H), 4.11-4.02 (m, 3H, 6′-Ha, 3″-H, 6″-Ha), 3.94 (dd, J=8.8, 8.3 Hz, 1H, 3-H), 3.92-3.84 (m, 3H, 6-Hb, 6″-Hb, 6′″-Ha), 3.75-3.64 (m, 6H, 4-H, 3′-H, 6′-Hb, 4′″-H, 5″-H, 6″-Hb), 3.59 (ddd, J=9.3, 4.2, 2.0 Hz, 1H, 5-H), 3.57-3.53 (m, 2H, 4″-H, 3′″-H), 3.46-3.50 (m, 1H, 5′-H), 3.18-3.14 (m, 2H, 2′-H, 2′″-H), 2.32 (s, 3H, SPhCH3), 1.79 (s, 3H, Ac), 1.68 (s, 3H, Ac), 1.04 (s, 9H, tBu), 0.99 (s, 9H, tBu); 13C NMR (150 MHz, CDCl3): δ 170.2 (C), 170.1 (C), 165.5 (C), 165.1 (C), 138.3 (C), 137.32 (C), 137.31 (C), 137.1 (C), 137.0 (C), 135.9 (CH), 135.60 (CH), 135.58 (CH), 135.5 (CH), 133.7 (CH), 133.4 (CH), 133.2 (CH), 132.9 (C), 132.8 (CH), 132.7 (C), 131.4 (C), 131.1 (CH), 129.93 (CH), 129.91 (CH), 129.84 (CH), 129.83 (CH), 129.7 (CH), 129.63 (CH), 129.60 (CH), 129.5 (CH), 129.0 (CH), 128.6 (CH), 128.5 (CH), 128.43 (CH), 128.14 (C), 128.09 (CH), 127.84 (CH), 127.78 (CH), 127.74 (CH), 127.63 (CH), 127.56 (CH), 127.4 (CH), 121.7 (C), 121.1 (C), 98.3 (CH), 97.4 (CH), 97.0 (CH), 85.9 (CH), 84.8 (CH), 79.9 (CH), 78.5 (CH), 76.4 (CH), 74.6 (CH2), 74.2 (CH2), 74.1 (CH2), 73.7 (CH), 73.1 (CH), 72.93 (CH), 72.91 (CH), 72.88 (CH2), 72.6 (CH), 72.3 (CH), 71.9 (CH), 68.8 (CH), 65.3 (CH), 63.7 (CH2), 63.5 (CH), 63.0 (CH), 62.5 (CH2), 62.2 (CH2), 62.1 (CH2), 26.9 (CH3), 26.8 (CH3), 21.1 (CH3), 20.7 (CH3), 20.4 (CH3), 19.4 (C), 19.2 (C); HRMS (MALDI): m/z calcd for C109H116Br2N6O22SSi2Na ([M+Na]+): 2133.1855, found: 2133.1912.
  • N-Benzyl-N-benzyloxycarbonyl-5-aminopentyl [2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyran osyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzyl-α-L-idopyranosyl)-(1→4)-[2-azid o-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-( 1→4)-6-O-acetyl-2-O-benzoyl-3-O-benzyl-β-D-glucopyranoside (12)
  • A solution of the thioglycoside 11 (0.78 g, 0.37 mmol) and N-benzyl-N-benzyloxycarbonyl-5-aminopentanol4 (0.12 g, 0.37 mmol) in CH2Cl2 (10 mL) with freshly dried AW-300 molecular sieves (1 g) was stirred at room temperature for 1 h under N2. The reaction flask was cooled to −78° C., NIS (0.08 g) and TfOH (27 μmL) were added to the solution, and the mixture was gradually warmed up to room temperature. After the thioglycoside was consumed completely according to TLC plate analysis (2 h), Et3N was added to quench the reaction, the mixture was filtered through Celite, and the solid was washed by CH2C12. The filtrate was washed with 10% Na2S2O3(aq), dried over MgSO4 and concentrated under reduced pressure to furnish a residue, which was purified by flash column chromatography (ethyl acetate/hexane=1/3) to give the expected tetrasaccharide 12 (0.76 g, 89%). IR (thin film): v 2926, 2108, 1737, 1266, 1112, 701 cm−1; 1H NMR (600 MHz, CDCl3): δ 8.03-7.97 (m, 4H, Bz-H), 7.67-7.62 (m, 4H, Ar—H), 7.61-7.55 (m, 4H, Ar—H), 7.50-7.39 (m, 6H, Ar—H), 7.38-7.26 (m, 23H, Ar—H), 7.26-7.20 (m, 7H, Ar—H), 7.20-7.15 (m, 6H, Ar—H), 7.11-7.05 (m, 5H, Ar—H), 5.48 (d, J=3.7 Hz, 1H, 1′-H), 5.42 (s, 1H, 1″-H), 5.25 (t, J=8.3 Hz, 1H, 2-H), 5.19 (s, 1H, 2″-H), 5.12 (d, J=7.4 Hz, 2H, ArCH2), 4.91 (d, J=11.6 Hz, 1H, ArCH2), 4.84 (d, J=11.2 Hz, 1H, ArCH2), 4.75-4.65 (m, 4H, ArCH2, 1″-H), 4.53-4.43 (m, 3H, ArCH2, 1-H), 4.41-4.32 (m, 4H, ArCH2, 6-Ha, 5″-H), 4.21-4.14 (m, 2H, ArCH2, 4′-H), 4.11-4.02 (m, 3H, 6′-Ha, 3″-H, 6″-Ha), 3.99-3.88 (m, 3H, 3-H, 6-Hb, 6′″-Ha), 3.85 (d, J=12.0 Hz, 6″-Hb), 3.77 (t, J=9.0 Hz, 1H, 4-H), 3.75-3.66 (m, 5H, 3′-H, 6′-Hb, 4′″-H, 5′″-H, 6″-Hb), 3.66-3.61 (m, 1H), 3.58-3.48 (m, 4H, 5-H, 5′-H, 3′″-H), 3.39-3.26 (m, 1H), 3.18-3.14 (m, 2H, 2′-H, 2′″-H), 3.08-2.89 (m, 2H), 2.46 (d, J=2.6 Hz, 1H, 4″-OH), 1.79 (s, 3H, Ac), 1.66-1.59 (m, 2H), 1.63 (s, 3H, Ac), 1.50-1.25 (m, 4H), 1.03 (s, 9H, tBu), 0.98 (s, 9H, tBu); 13C NMR (150 MHz, CDCl3): δ 170.2 (C), 170.1 (C), 165.5 (C), 165.0 (C), 156.6/156.0 (C), 137.9 (C), 137.8 (C), 137.4 (C), 137.3 (C), 137.1 (C), 137.0 (C), 136.8/136.7 (C), 135.9 (CH), 135.6 (CH), 135.56 (CH), 135.52 (CH), 133.3 (C), 133.2 (CH), 132.9 (C), 132.8 (C), 132.7 (C), 131.4 (CH), 131.1 (CH), 129.93 (CH), 129.91 (CH), 129.8 (CH), 129.7 (CH), 129.62 (CH), 129.60 (CH), 128.9 (CH), 128.56 (CH), 128.51 (CH), 128.45 (CH), 128.3 (CH), 128.1 (CH), 127.85 (CH), 127.78 (CH), 127.72 (CH), 127.61 (CH), 127.56 (CH), 127.4 (CH), 127.23 (CH), 127.17 (CH), 127.09 (CH), 121.6 (C), 121.1 (C), 100.8 (CH), 98.2 (CH), 97.2 (CH), 96.9 (CH), 83.3 (CH), 79.9 (CH), 78.5 (CH), 74.3 (CH2), 74.2 (CH2), 74.10 (CH2), 73.96 (CH), 73.7 (CH), 73.1 (CH), 72.93 (CH), 72.88 (CH2), 72.86 (CH), 72.6 (CH), 72.5 (CH2), 72.3 (CH), 71.9 (CH), 69.7 (CH2), 69.6 (CH2), 68.8 (CH), 67.0 (CH2), 65.3 (CH), 63.6 (CH2), 63.5 (CH), 63.0 (CH), 62.5 (CH2), 62.2 (CH2), 62.1 (CH2), 53.7 (CH2), 50.4 (CH2), 50.1 (CH2), 47.0 (CH2), 46.0 (CH2), 21.7 (CH3), 29.2 (CH3), 28.9 (CH2), 27.6/27.2 (CH2), 26.9 (CH3), 26.8 (CH3), 23.0 (CH2), 20.7 (CH3), 20.3 (CH3), 19.4 (C), 19.2 (C); HRMS (ESI): m/z calcd for C122H133Br2N7O25Si2Na ([M+Na]+): 2332.7154, found: 2332.7119.
  • N-Benzyl-N-benzyloxycarbonyl-5-aminopentyl [2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-4-O-(2-naphthyl methyl)-α-D-glucopyranosyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzyl-α-L-idop yranosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzyl-β-D-glucopyrano syl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-(6-O-acetyl-2-O-benzoyl-3-O-benzyl-α-L-idopyranosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucop yranosyl]-(1→4)-6-O-acetyl-2-O-benzoyl-3-O-benzyl-β-D-glucopyranoside (13)
  • A solution of the thioglycoside 10 (1.4 equiv.) and 4′″-alcohol 12 (1.0 equiv.) in anhydrous CH2C12 (10 mL/total grams of the thioglycoside and the 4″-alcohol) with freshly activated AW-300 molecular sieves (0.5 g/total grams of the thioglycoside and the 4′″-alcohol) was stirred at room temperature for 1 h and then cooled to −40° C., followed by the addition of NIS (1.6 equiv.). After stirring for 5 min, TfOH (0.4 equiv.) was added to the solution slowly through a microsyringe. The reaction was warmed up to room temperature gradually and kept stirring until the complete consumption of the thioglycoside by TLC plate analysis (about 2 h). The mixture was neutralized by addition of Et3N, diluted with CH2Cl2 and filtered through Celite. The resulting solution was washed with 10% Na2S2O3(aq) and H2O. The organic layer was combined, dried over MgSO4 and concentrated under reduced pressure. The residue was purified by flash column chromatography to yield octasaccharides 13 (72%).
  • [α]26 D —2.3 (c 5.0, CHCl3); IR (thin film): v 2928, 2108, 1741, 1698, 1264, 1112, 1038, 701 cm−1; 1H NMR (600 MHz, CDCl3): δ 8.02-7.97 (m, 4H, Bz-H), 7.96-7.92 (m, 2H, Bz-H), 7.85-7.82 (m, 1H, Ar—H), 7.81-7.78 (m, 2H, Ar—H), 7.76-7.71 (m, 3H, Ar—H), 7.69-7.64 (m, 4H, Ar—H), 7.63-7.60 (m, 2H, Ar—H), 7.58-7.47 (m, 15H, Ar—H), 7.44-7.40 (m, 2H, Ar—H), 7.40-7.33 (m, 14H, Ar—H), 7.33-7.18 (m, 44H, Ar—H), 7.18-7.09 (m, 12H, Ar—H), 7.07-6.97 (m, 8H, Ar—H), 5.46 (d, J=4.0 Hz, 1H, 1′-H), 5.44 (d, J=4.0 Hz, 1H, 1′″″-H), 5.41 (s, 1H, 1″-H), 5.29-5.21 (m, 3H, 2-H, 2″″-H, 1″″″-H), 5.18 (bs, 1H, 2″-H), 5.15 (dd, J=3.4, 2.8 Hz, 1H, 2″″″-H), 5.12 (d, J=7.9 Hz, 2H, PhCH2 in Cbz), 4.91-4.84 (m, 4H, 1″″-H, ArCH2), 4.83 (s, 2H, NaphCH2), 4.79 (d, J=11.1 Hz, 1H, ArCH2), 4.74-4.66 (m, 6H, 1′″-H, ArCH2), 4.60 (d, J=3.5 Hz, 1H, 1′″″″-H), 4.53 (d, J=10.1 Hz, 1H, ArCH2), 4.49-4.42 (m, 3H, 1-H, ArCH2), 4.40-4.32 (m, 5H), 4.18-4.04 (m, 8H), 4.04-3.99 (m, 3H), 3.97 (t, J=3.8 Hz, 1H, 3″″″-H), 3.92 (t, J=8.8 Hz, 1H, 3-H), 3.90-3.73 (m, 11H), 3.73-3.63 (m, 7H), 3.63-3.55 (m, 2H), 3.55-3.48 (m, 3H), 3.46-3.42 (m, 2H), 3.40-3.27 (m, 3H, 5″″-H-5, 5′″″″-H, CH2 in linker), 3.27 (dd, J=10.2, 3.5 Hz, 1H, 2′″-H), 3.15-3.10 (m, 2H, 2′-H, 2′″″-H), 3.08-2.89 (m, 2H, CH2 in linker), 1.70 (s, 3H, Ac), 1.59 (bs, 5H, Ac, CH2 in linker), 1.46 (s, 3H, Ac), 1.38 (s, 3H, Ac), 1.49-1.25 (m, 4H, CH2 in linker), 1.10 (s, 9H, tBu), 1.04 (s, 9H, tBu), 0.93 (s, 18H, tBu×2); 13C NMR (150 MHz, CDCl3): δ 170.11 (C), 170.09 (C), 170.0 (C), 169.4 (C), 165.54(C), 165.46 (C), 165.0 (C), 164.8 (C), 156.6/156.0 (C), 137.94 (C), 137.90 (C), 137.8 (C), 137.6 (C), 137.40 (C), 137.36 (C), 137.19 (C), 137.15 (C), 137.1 (C), 136.9 (C), 136.8 (C), 136.0 (CH), 135.89 (CH), 135.86 (CH), 135.8 (CH), 135.62 (CH), 135.56 (CH), 135.52 (CH), 135.45 (C), 133.7 (C), 133.4 (CH), 133.34 (CH), 133.27 (C), 133.2 (CH), 133.20 (C), 133.18 (C), 133.1 (CH), 133.0 (C), 132.9 (C), 132.8 (C), 132.2 (C), 131.4 (CH), 131.1 (CH), 131.04 (CH), 131.03 (CH), 130.2 (CH), 130.0 (CH), 129.83 (CH), 129.77 (CH), 129.7 (CH), 129.6 (CH), 129.5 (CH), 129.4 (CH), 129.15 (CH), 129.09 (CH), 129.01 (CH), 128.96 (C), 128.9 (CH), 128.8 (CH), 128.6 (CH), 128.54 (CH), 128.46 (CH), 128.4 (CH), 128.3 (CH), 128.2 (CH), 128.13 (CH), 128.10 (CH), 128.08 (CH), 127.9 (CH), 127.8 (CH), 127.72 (CH), 127.69 (CH), 127.65 (CH), 127.61 (CH), 127.59 (CH), 127.56 (CH), 127.5 (CH), 127.43 (CH), 127.38 (CH), 127.24 (CH), 127.18 (CH), 127.1 (CH), 126.4 (CH), 126.2 (CH), 126.0 (CH), 125.7 (CH), 125.3 (CH), 121.6 (C), 121.1 (C), 121.0 (C), 100.8 (CH), 99.8 (CH), 98.5 (CH), 98.3 (CH), 97.4 (CH), 97.3 (CH), 96.99 (CH), 96.96 (CH), 83.6 (CH), 83.3 (CH), 80.4 (CH), 78.4 (CH), 78.2 (CH), 78.0 (CH), 77.7 (CH), 76.0 (CH), 75.1 (CH2), 74.7 (CH2), 74.4 (CH), 74.3 (CH2), 74.2 (CH2), 74.0 (CH/CH3), 73.9 (CH), 73.5 (CH), 73.4 (CH), 73.2 (CH), 73.1 (CH/CH2), 72.94 (CH), 72.85 (CH), 72.83 (CH2), 72.77 (CH), 72.6 (CH), 72.4 (CH), 72.2 (CH), 69.7/69.6 (CH2), 69.1 (CH), 68.5 (CH), 67.1 (CH2), 65.5 (CH), 64.8 (CH), 64.0 (CH), 63.5 (CH), 63.4 (CH), 63.1 (CH), 62.7 (CH2), 62.4 (CH2), 62.3 (CH2), 62.0 (CH2), 61.9 (CH2), 61.4 (CH2), 60.9 (CH2), 50.4/50.1 (CH2), 47.0/46.0 (CH2), 29.0 (CH2), 27.6/27.2 (CH2), 26.93 (CH3), 26.87 (CH3), 26.81 (CH3), 26.75 (CH3), 26.7 (CH3), 23.0 (CH2), 21.4 (CH3), 20.7 (CH3), 20.34 (CH3), 20.32 (CH3), 19.9 (CH3), 19.42 (C), 19.37 (C), 19.32 (C), 19.28 (C); HRMS (MALDI): m/z calcd for C235H249Br4N13O47Si4Na ([M+Na]+): 4462.5806, found: 4462.5928.
  • N-Benzyl-N-benzyloxycarbonyl-5-aminopentyl [2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-4-O-(2-naphthyl methyl)-α-D-glucopyranosyl]-(1→4)-(3-O-benzyl-α-L-idopyranosyl)-(1→4)-[2-azid o-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-(3-O-benzyl-β-D-glucopyranosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-t ert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-(3-O-benzyl-α-L-idopyr anosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-3-O-benzyl-β-D-glucopyranoside (14)
  • NaOMe (5 equiv. per acyl group) was added to a solution of the octasaccharide 13 (1.68 g, 0.38 mmol) in a mixed solvent (MeOH/CH2Cl2=1/1) at room temperature under N2 atmosphere. After stirring for 16 h, the reaction was neutralized by adding DOWEX 50WX4-200 resin. The mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by flash column chromatography to obtain the deacylated compound 14 (1.23 g, 85%). [α]27D+8.6 (c 5.0, CHCl3); IR (thin film): v 3476, 2929, 2109, 1684, 1254, 1070, 1030, 700 cm−1; 1H NMR (600 MHz, CDCl3): δ 7.83-7.80 (m, 1H, Ar—H), 7.78-7.711 (m, 2H, Ar—H), 7.71-7.68 (m, 4H, Ar—H), 7.68-7.65 (m, 2H, Ar—H), 7.61-7.59 (s, 1H, Ar—H), 7.49-7.47 (m, 2H, Ar—H), 7.45-7.33 (m, 39H, Ar—H), 7.33-7.27 (m, 13H, Ar—H), 7.27-7.25 (m, 3H, Ar—H), 7.23-7.18 (m, 5H, Ar—H), 7.12-7.16 (m, 7H, Ar—H), 7.04 (d, J=8.2 Hz, 2H, Ar—H), 7.00 (d, J=8.2 Hz, 2H, Ar—H), 5.64 (b s, 1H), 5.61 (d, J=3.8 Hz, 1H), 5.23 (s, 1H), 5.21-5.13 (m, 3H), 5.09 (d, J=10.7 Hz, 1H), 5.05 (d, J=3.7 Hz, 1H), 5.01 (d, J=4.1 Hz, 1H), 4.95-4.87 (m, 3H), 4.84-4.81 (m, 3H), 4.79-4.74 (m, 3H), 4.68-4.55 (m, 5H), 4.54-4.46 (m, 5H), 4.34-4.24 (m, 1H), 4.19-4.14 (m, 2H), 4.13-4.10 (m, 1H), 4.10-4.05 (m, 1H), 4.05-3.99 (m, 2H), 3.99-3.92 (m, 2H), 3.92-3.89 (m, 2H), 3.87-3.80 (m, 10H), 3.77-3.72 (m, 3H), 3.71-3.64 (m, 5H), 3.62-3.51 (m, 11H), 3.48-3.39 (m, 4H), 3.38-3.36 (m, 1H), 3.34-3.16 (m, 7H), 3.11-3.06 (m, 1H, CH2 in linker), 3.05-2.99 (m, 1H, CH2 in linker), 1.66-1.49 (m, 6H, CH2 in linker), 1.10 (s, 9H, tBu), 1.08 (s, 9H, tBu), 1.05 (s, 9H, tBu), 1.04 (s, 9H, tBu); 13C NMR (150 MHz, CDCl3): δ 156.7/156.3 (C), 138.4 (C), 137.7 (C), 137.5 (C), 137.4 (C), 136.8 (C), 136.7 (C), 136.6 (C), 136.00 (CH), 135.97 (CH), 135.91 (CH), 135.87 (CH), 135.8 (CH), 135.7 (CH), 135.61 (CH), 135.57 (CH), 135.3 (C), 133.33 (C), 133.28 (C), 133.2 (C), 133.1 (C), 133.0 (C), 132.9 (C), 131.51 (CH), 131.47 (CH), 131.2 (CH), 129.9 (CH), 129.8 (CH), 129.71 (CH), 129.67 (CH), 129.6 (CH), 129.4 (CH), 129.3 (CH), 129.2 (CH), 128.52 (CH), 128.47 (CH), 128.4 (CH), 128.35 (CH), 128.31 (CH), 128.21 (CH), 128.18 (CH), 127.90 (CH), 127.85 (CH), 127.81 (CH), 127.76 (CH), 127.70 (CH), 127.68 (CH), 127.6 (CH), 127.5 (CH), 127.3 (CH), 127.2 (CH), 126.2 (CH), 126.1 (CH), 125.9 (CH), 125.5 (CH), 121.8 (C), 121.7 (C), 121.5 (C), 102.9/102.7 (CH), 102.1 (CH), 99.9 (CH), 99.8 (CH), 97.3 (CH), 97.1 (CH), 94.8 (CH), 94.2 (CH), 84.9 (CH), 84.6 (CH), 81.1 (CH), 79.4 (CH), 78.9 (CH), 78.6 (CH), 77.9 (CH), 77.2 (CH), 76.1 (CH), 75.8 (CH), 75.2 (CH), 75.1 (CH2), 74.9 (CH2), 74.74 (CH/CH2), 74.66 (CH2), 74.6 (CH), 74.3 (CH2), 73.9 (CH2), 73.0 (CH), 72.90 (CH), 72.85 (CH), 72.6 (CH), 72.4 (CH), 72.3 (CH/CH2), 72.2 (CH2), 72.0 (CH), 70.4 (CH), 70.0 (CH2), 69.9 (CH), 67.2 (CH2), 66.8 (CH), 66.7 (CH), 66.4 (CH), 66.2 (CH), 63.8 (CH), 63.6 (CH), 63.34 (CH), 63.31 (CH), 62.4 (CH2), 62.3 (CH2), 62.1 (CH2), 62.0 (CH2), 61.5 (CH2), 61.31 (CH2), 61.26 (CH2), 53.7 (CH2), 50.4/50.2 (CH2), 46.9/45.9 (CH2), 29.6 (CH2), 29.2 (CH2), 28.8 (CH2), 27.7 (CH2), 26.88 (CH3), 26.86 (CH3), 26.84 (CH3), 23.1 (CH2), 19.42 (C), 19.38 (C), 19.31 (C); HRMS (MALDI): m/z calcd for C199H225Br4N13O39Si4Na ([M+Na]+): 3877.9968, found: 3877.9971.
  • N-Benzyl-N-benzyloxycarbonyl-5-aminopentyl [2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-4-O-(2-naphthyl methyl)-α-D-glucopyranosyl]-(1→4)-(methyl 3-O-benzyl-α-L-idopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-te rt-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-(methyl 3-O-benzyl-β-D-glucopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-tert-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-(methyl 3-O-benzyl-α-L-idopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-6-O-te rt-butyldiphenylsilyl-2-deoxy-α-D-glucopyranosyl]-(1→4)-methyl 3-O-benzyl-β-D-glucopyranosiduronate (15)
  • Bis(acetoxy)iodobenzene (BAIB, 331 mg, 1.03 mmol) was added to a solution of the compound 14 (330 mg, 0.09 mmol) in a mixed solvent (CH2Cl2/H2O=2/1, 3 mL) at room temperature. After 5 min, 2,2,6,6-tetramethyl-1-piperidinyloxyl free radical (TEMPO, 7 mg, 0.04 mmol) was placed into the mixture, which was, then, kept stirring for 8 h. To quench the reaction, 10% Na2S2O3(aq) (3 mL) was added, and the desired material was extracted with CH2C12. The combined organic layer was washed with brine, dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. To a solution of this residue in THF (2.5 mL) was then added LiOH (0.5 M solution in water, 1.7 mL, 0.9 mmol) at room temperature. After 4 h, the reaction was neutralized by adding DOWEX 50WX-200 resin, filtered and concentrated under reduced pressure. To the crude uronate solution in CH2Cl2 (3 mL) was added an ethereal solution of CH2N2 (3 mL, 1.28 mmol), and the reaction mixture was stirred at room temperature for 12 h. The reaction solution was quenched by acetic acid and the resulting mixture was concentrated under reduced pressure to yield the crude residue. Purification by flash column chromatography (ethyl acetate/hexane=1/5) gave compound 42 (220 mg, 65% overall). [α]22 D+8.6 (c 4.0, CHCl3); IR (thin film): v 3514, 2931, 2110, 1754, 1215, 1112, 1013, 701 cm−1; 1H NMR (600 MHz, CDCl3): δ 7.88-7.80 (m, 1H, Ar—H), 7.75-7.66 (m, 16H, Ar—H), 7.65-7.62 (m, 2H, Ar—H), 7.59-7.57 (m, 1H, Ar—H), 7.50-7.44 (m, 5H, Ar—H), 7.40-7.31 (m, 38H, Ar—H), 7.31-7.23 (m, 23H, Ar—H), 7.18-7.14 (m, 1H, Ar—H), 7.14-7.11 (m, 2H, Ar—H), 7.10-7.06 (m, 2H, Ar—H), 7.05-7.02 (m, 2H, Ar—H), 5.54-5.49 (m, 2H), 5.38 (s, 2H), 5.20-5.14 (m, 2H), 5.05-4.99 (m, 2H), 4.99-4.91 (m, 3H), 4.90-4.85 (m, 1H), 4.82-4.73 (m, 6H), 4.69-4.62 (m, 3H), 4.61-4.53 (m, 4H), 4.51-4.45 (m, 5H), 4.30-4.21 (m, 1H), 4.16-4.09 (m, 2H), 4.09-4.03 (m, 2H), 4.01-3.92 (m, 5H), 3.91-3.84 (m, 7H), 3.83-3.77 (m, 6H), 3.75-3.71 (m, 3H), 3.70-3.65 (m, 3H), 3.63-3.57 (m, 1H), 3.56-3.50 (m, 2H), 3.50-3.45 (m, 3H), 3.42-3.30 (m, 9H), 3.27-3.22 (m, 6H), 3.19-3.13 (m, 5H), 1.74-1.45 (m, 6H), 1.10 (s, 9H, tBu), 1.073 (s, 9H, tBu), 1.070 (s, 9H, tBu), 1.06 (s, 9H, tBu); 13C NMR (150 MHz, CDCl3): δ 169.0 (C), 168.6 (C), 168.1 (C), 156.7/156.3 (C), 138.3 (C), 138.23 (C), 138.16 (C), 137.8 (C), 137.14 (C), 137.12 (C), 137.0 (C), 136.92 (C), 136.86 (C), 136.8 (C), 136.7 (C), 136.60 (C), 136.57 (C), 136.0 (CH), 135.9 (CH), 135.82 (CH), 135.79 (CH), 135.6 (CH), 135.54 (CH), 135.49 (CH), 133.5 (C), 133.4 (C), 133.3 (C), 133.21 (C), 133.18 (C), 133.10 (C), 133.08 (C), 133.0 (C), 132.93 (C), 132.90 (C), 132.87 (C), 132.75 (C), 131.5 (CH), 131.4 (CH), 131.11 (CH), 131.08 (CH), 130.4 (CH), 130.0 (CH), 129.9 (CH), 129.74 (CH), 129.72 (CH), 129.68 (CH), 129.65 (CH), 129.63 (CH), 129.60 (CH), 129.1 (CH), 129.0 (CH), 128.61 (CH), 128.58 (C), 128.5 (CH), 128.4 (CH), 128.34 (CH), 128.29 (CH), 128.21 (CH), 128.17 (CH), 128. (CH), 128.0 (CH), 127.9 (CH), 127.84 (CH), 127.81 (CH), 127.71 (CH), 127.66 (CH), 127.55 (CH), 127.53 (CH), 127.32 (CH), 127.28 (CH), 127.17 (CH), 126.2 (CH), 125.9 (CH), 125.8 (CH), 125.2 (CH), 121.8 (C), 121.7 (C), 121.08 (C), 121.06 (C), 103.1 (CH), 102.8 (CH), 102.2 (CH), 101.0 (CH), 100.8 (CH), 97.20 (CH), 97.15 (CH), 95.4 (CH), 95.2 (CH), 84.2 (CH), 84.0 (CH), 80.9 (CH), 79.2 (CH), 78.3 (CH), 78.1 (CH), 77.6 (CH), 76.1 (CH), 75.1 (CH2), 74.92 (CH2), 74.89 (CH3), 74.87 (CH2), 74.8 (CH), 74.7 (CH2), 74.6 (CH), 74.5 (CH2), 74.4 (CH2), 74.2 (CH), 74.0 (CH), 73.7 (CH), 73.6 (CH), 73.1 (CH2), 72.9 (CH2), 72.7 (CH), 72.55 (CH), 72.49 (CH2), 72.4 (CH/CH2), 72.3 (CH), 72.2 (CH2), 72.1 (CH), 72.0 (CH), 71.4 (CH), 70.4/70.0 (CH2), 67.4 (CH), 67.2 (CH2), 67.1 (CH), 66.9 (CH), 66.6 (CH), 63.7 (CH), 63.2 (CH), 63.0 (CH), 62.1 (CH2), 61.9 (CH2), 61.1 (CH2), 53.8 (CH2), 52.3 (CH3), 52.1 (CH3), 51.7 (CH3), 51.6 (CH3), 50.4/50.2 (CH2), 46.9/45.9 (CH2), 31.9 (CH2), 29.7 (CH2), 29.6 (CH2), 29.3 (CH2), 26.92 (CH3), 29.0/28.8 (CH2), 27.0 (CH3), 26.94 (CH3), 26.89 (CH3), 23.3 (CH2), 23.0 (CH2), 22.70 (CH2), 22.67 (CH2), 22.6 (CH2), 19.42 (C), 19.41 (C), 19.37 (C); HRMS (MALDI): m/z calcd for C203H225Br4N13O43Si4Na ([M+Na]+): 3990.0383, found: 3990.0508.
  • N-Benzyl-N-benzyloxycarbonyl-5-aminopentyl [2-azido-3-O-(4-bromobenzyl)-2-deoxy-4-O-(2-naphthylmethyl)-α-D-glucopyranos yl]-(1→4)-(methyl 3-O-benzyl-α-L-idopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-2-deo xy-α-D-glucopyranosyl]-(1→4)-(methyl 3-O-benzyl-β-D-glucopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-2-de oxy-α-D-glucopyranosyl]-(1→4)-(methyl 3-O-benzyl-α-L-idopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-2-deo xy-α-D-glucopyranosyl]-(1→4)-methyl 3-O-benzyl-β-D-glucopyranosiduronate (16)
  • A 70% solution of HF in pyridine (500 equiv.) was added to an ice-cooled solution of the compound 15 (210 mg, 0.05 mmol) in a mixed solvent (pyridine/THF=1/1, v/v, 40 mL/g of octasaccharide). The mixture was allowed to warm to room temperature and stirred for 3 d. The solution was concentrated under reduced pressure, and the residue was purified by flash column chromatography to afford the compound 16 (135 mg, 83%). [α]27 D —29.7 (c 2.0, CHCl3); IR (thin film): v 2924, 2109, 1748, 1027, 699 cm−1; 1H NMR (600 MHz, CDCl3): δ 7.81-7.79 (m, 1H, Ar—H), 7.78-7.74 (m, 2H, Ar—H), 7.63 (bs, 1H, Ar—H), 7.48-7.45 (m, 2H, Ar—H), 7.43-7.41 (m, 2H, Ar—H), 7.38-7.34 (m, 18H, Ar—H), 7.33-7.30 (m, 8H, Ar—H), 7.30-7.26 (m, 7H, Ar—H), 7.25-7.22 (m, 4H, Ar—H), 7.20-7.18 (m, 2H, Ar—H), 7.16-7.13 (m, 1H, Ar—H), 7.10-7.07 (m, 2H, Ar—H), 7.04-7.00 (m, 4H, Ar—H), 5.56 (bs, 1H), 5.47 (d, J=3.5 Hz, 1H), 5.23 (d, J=8.5 Hz, 2H), 5.19-5.12 (m, 2H), 5.11-5.03 (m, 1H), 4.99-4.95 (m, 2H), 4.92 (d, J=10.9 Hz, 1H), 4.89 (d, J=3.5 Hz, 1H), 4.86 (d, J=11.8 Hz, 1H), 4.81-4.77 (m, 3H), 4.74-4.72 (m, 3H), 4.71 (d, J=5.0 Hz, 1H), 4.69 (d, J=5.0 Hz, 1H), 4.61-4.56 (m, 3H), 4.56-4.50 (m, 3H), 4.49-4.42 (m, 4H), 4.31-4.24 (m, 1H), 4.07 (t, J=9.0 Hz, 1H), 4.03 (t, J=9.0 Hz, 1H), 4.00 (bs, 1H), 3.98-3.93 (m, 3H), 3.92-3.85 (m, 4H), 3.85-3.81 (m, 3H), 3.81-3.78 (m, 2H), 3.78-3.75 (m, 3H), 3.73-3.69 (m, 8H), 3.68-3.65 (m, 3H), 3.65-3.62 (m, 3H), 3.62-3.59 (m, 3H), 3.58-3.55 (m, 2H), 3.51 (s, 3H), 3.50-3.49 (m, 1H), 3.49-3.46 (m, 1H), 3.45-3.43 (m, 5H), 3.42-3.41 (m, 1H), 3.40-3.37 (m, 4H), 3.24-3.18 (m, 3H), 1.34-1.25 (m, 6H, CH2 in linker); 13C NMR (150 MHz, CDCl3): δ 169.5 (C), 169.4 (C), 168.8 (C), 168.5 (C), 156.7/156.3 (C), 138.12 (C), 138.06 (C), 138.0 (C), 137.7 (C), 137.0 (C), 136.8 (C), 136.72 (C), 136.65 (C), 136.52 (C), 136.47 (C), 135.0 (C), 133.1 (C), 132.9 (C), 131.45 (CH), 131.37 (CH), 131.14 (CH), 131.10 (CH), 129.52 (CH), 129.46 (CH), 129.0 (CH), 128.9 (CH), 128.6 (CH), 128.5 (CH), 128.40 (CH), 128.36 (CH), 128.3 (CH), 127.9 (CH), 127.8 (CH), 127.7 (CH), 127.6 (CH), 127.2 (CH), 127.1 (CH), 126.28 (CH), 126.27 (CH), 126.1 (CH), 125.4 (CH), 121.7 (C), 121.5 (C), 121.14 (C), 121.09 (C), 103.2 (CH), 103.1 (CH), 102.9 (CH), 100.9 (CH), 100.7 (CH), 97.4 (CH), 97.2 (CH), 96.0 (CH), 95.4 (CH), 83.9 (CH), 80.7 (CH), 79.3 (CH), 78.4 (CH), 78.3 (CH), 76.2 (CH), 75.0 (CH2), 74.9 (CH2), 74.8 (CH2), 74.73 (CH2), 74.66 (CH), 74.6 (CH3), 74.5 (CH), 74.1 (CH), 73.9 (CH), 73.50 (CH2), 73.47 (CH2), 73.1 (CH), 72.9 (CH), 72.6 (CH2), 72.5 (CH2), 72.2 (CH), 72.1 (CH), 71.9 (CH), 71.6 (CH2), 71.0 (CH2), 70.4 (CH2), 70.3 (CH2), 70.0 (CH2), 67.7 (CH), 67.5 (CH), 67.2 (CH3/CH2), 67.1 (CH), 66.8 (CH), 63.5 (CH), 63.4 (CH), 63.3 (CH), 61.8 (CH2), 61.0 (CH2), 60.9 (CH2), 60.3 (CH2), 52.7 (CH3), 52.6 (CH3), 52.14 (CH3), 52.13 (CH3), 50.4/50.2 (CH2), 46.9/45.9 (CH2), 31.9 (CH2), 31.6 (CH2), 30.0 (CH3), 29.63 (CH2), 29.59 (CH2), 29.3 (CH2), 29.0 (CH2), 28.7 (CH2), 27.6 (CH2), 27.1 (CH2), 27.0 (CH2), 23.2 (CH2), 23.0 (CH2), 22.6 (CH2), 19.2 (C), 14.1 (CH2), 13.9 (C); HRMS (MALDI): m/z calcd for C139H153Br4N13O43Na [M+Na]+: 3036.4155, found: 3036.4241.
  • N-Benzyl-N-benzyloxycarbonyl-5-aminopentyl [2-azido-3-O-(4-bromobenzyl)-2-deoxy-4-O-(2-naphthylmethyl)-6-O-sulfonato-α-D-glucopyranosyl)-(1→4)-(methyl 3-O-benzyl-2-O-sulfonato-α-L-idopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromo benzyl)-2-deoxy-6-O-sulfonato-α-D-glucopyranosyl]-(1→4)-(methyl 3-O-benzyl-2-O-sulfonato-β-D-glucopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bro mobenzyl)-2-deoxy-6-O-sulfonato-α-D-glucopyranosyl)-(1→4)-(methyl 3-O-benzyl-2-O-sulfonato-α-L-idopyranosyluronate)-(1→4)-[2-azido-3-O-(4-bromo benzyl)-2-deoxy-6-O-sulfonato-α-D-glucopyranosyl)-(1→4)-methyl 3-O-benzyl-2-O-sulfonato-β-D-glucopyranosiduronate octasodium salt (17)
  • A solution of compound 16 (190 mg, 0.06 mmol) and SO3.Et3N (4 equiv. per OH group of the octasaccharide) in DMF (10 mL/g of octasaccharide) was kept stirring at 60° C. under N2 for 1 d. The reaction flask was cooled down to room temperature, a solution of phosphate buffer (pH=7, 2.0 mL) was added and the mixture was kept stirring for another 30 min. The resulting solution was concentrated in vacuo, and the residue was purified by Sephadex LH-20 with MeOH as eluent to get the O-sulfonated compound 17 (226 mg, 94%). IR (thin film): v 3464, 2111, 1747, 1236, 1068, 1005, 701 cm−1; 1H NMR (600 MHz, CD3OD): δ 7.81-7.72 (m, 3H, Ar—H), 7.70 (bs, 1H, Ar—H), 7.55-7.49 (m, 6H, Ar—H), 7.46-7.37 (m, 18H, Ar—H), 7.36-7.33 (m, 5H, Ar—H), 7.32-7.26 (m, 9H, Ar—H), 7.25-7.21 (m, 4H, Ar—H), 7.21-7.17 (m, 3H, Ar—H), 7.04 (d, J=8.5 Hz, 2H, Ar—H), 7.01 (d, J=8.4 Hz, 2H, Ar—H), 5.56 (s, 1H), 5.48-5.42 (m, 3H), 5.32 (d, J=10.0 Hz, 1H), 5.19-5.09 (m, 5H), 5.04 (d, J=3.0 Hz, 1H), 4.99-4.94 (m, 2H), 4.93-4.91 (m, 1H), 4.85-4.77 (m, 8H, ArCH2), 4.73-4.70 (m, 2H), 4.69-4.68 (m, 1H), 4.67-4.63 (m, 2H), 4.59 (s, 2H), 4.55-4.48 (m, 4H), 4.45-4.38 (m, 3H), 4.37-4.31 (m, 4H), 4.30-4.25 (m, 3H), 4.25-4.20 (m, 5H), 4.15-4.10 (m, 2H), 4.06 (t, J=9.5 Hz, 1H), 4.04-3.99 (m, 2H), 3.97 (dd, J=8.5, 4.9 Hz, 1H), 3.91-3.86 (m, 4H), 3.86-3.83 (m, 1H), 3.80 (s, 3H), 3.76-3.71 (m, 2H), 3.71-3.64 (m, 6H), 3.64-3.59 (m, 3H), 3.55 (s, 3H), 3.48-3.46 (m, 1H), 3.45 (s, 3H), 3.43-3.41 (m, 1H), 3.41-3.38 (m, 1H), 3.30-3.27 (m, 2H), 3.26-3.18 (m, 2H), 1.64-1.50 (m, 4H, CH2 in linker), 1.41-1.29 (m, 2H, CH2 in linker); 13C NMR (150 MHz, CD3OD): δ 172.6 (C), 172.2 (C), 171.8 (C), 170.9 (C), 158.6/158.1 (C), 139.8 (C), 139.6 (C), 139.5 (C), 139.4 (C), 139.1 (C), 138.83 (C), 138.81 (C), 138.3 (C), 138.2 (C), 137.2 (C), 134.8 (C), 134.5 (C), 132.40 (CH), 132.37 (CH), 132.36 (CH), 132.2 (CH), 131.6 (CH), 130.7 (CH), 130.52 (CH), 130.46 (CH), 130.4 (CH), 129.3 (CH), 129.8 (CH), 129.73 (CH), 129.66 (CH), 129.6 (CH), 129.5 (CH), 129.29 (CH), 129.25 (CH), 129.11 (CH), 129.05 (CH), 129.0 (CH), 128.8 (CH), 128.7 (CH), 128.5 (CH), 128.4 (CH), 127.7 (CH), 127.2 (CH), 127.1 (CH), 126.9 (CH), 122.33 (C), 122.26 (C), 122.2 (C), 122.1 (C), 102.7 (CH), 101.8 (CH), 99.6 (C), 98.8 (CH), 98.6 (CH), 98.5 (CH), 98.4 (CH), 98.2 (CH), 84.1 (CH), 83.8 (CH), 81.7 (CH), 80.4 (CH), 80.3 (CH), 80.2 (CH), 80.0 (CH), 79.6 (CH), 78.9 (CH), 78.8 (CH), 77.0 (CH), 76.1 (CH2), 75.65 (CH2), 75.6 (CH2), 75.4 (CH2/CH), 75.2 (CH), 75.0 (CH), 74.9 (CH2), 74.8 (CH2), 74.2 (CH2), 73.6 (CH2), 73.5 (CH2), 73.4 (CH), 73.2 (CH), 72.5 (CH), 72.2 (C), 72.0 (C), 71.9 (C), 71.8 (CH), 71.5 (CH), 70.5 (CH), 70.32 (CH), 70.26 (CH2), 70.1 (CH2), 68.6 (CH2), 68.4 (CH2), 68.1 (CH), 68.0 (CH), 67.3 (CH2), 67.03 (CH2), 66.98 (CH2), 66.6 (CH2), 65.4 (CH), 65.0 (CH), 54.1 (CH), 53.6 (CH), 51.6 (CH2), 51.5 (CH2), 47.7 (CH2), 30.1 (CH2), 30.0 (CH2), 29.0 (CH2), 28.6 (CH2), 24.32 (CH2), 24.25 (CH2); LRMS (ESI): m/z calcd for C139H145Br4Ni3Na10O67S8 ([M−8H+10Na]2+): 1937.9, found: 1937.6.
  • O-sulfonato-α-D-glucopyranosyl]-(1→4)-(3-O-benzyl-2-O-sulfonato-β-D-glucuronopyranosyl)-(1→4)-[2-azido-3-O-(4-bromobenzyl)-2-deoxy-6-O-sulfonato-α-D-glucopyranosyl]-(1→4)-(3-O-benzyl-2-O-sulfonato-α-L-iduronopyranosyl)-(1→4)-[2-azido-3-O-(4-bromo-benzyl)-2-deoxy-6-O-sulfonato-α-D-glucopyranosyl]-(1→4)-3-O-benzyl-2-O-sulfonato-β-D-glucuronopyranoside dodecasodium salt (18)
  • CHCl3 (2.4 ml), MeOH (8.8 ml), NaOH (5 M solution in H2O, 1.2 mL) and H2O (1.2 mL) were added to the compound 17 (155 mg, 0.05 mmol) at room temperature. The mixture was kept stirring for 2 d, and the reaction solution was neutralized by 1 N HCl(aq). The mixture was concentrated in vacuo, and the residue was purified by gel permeation chromatography on Sephadex LH-20 using MeOH as eluent. The appropriate fractions were pooled and passed through a column of AG 50W-X8 Na+ resin with MeOH to get compound 18 (150 mg, 96%).
  • 1H NMR (600 MHz, CD3OD): δ 7.81-7.72 (m, 4H, Ar—H), 7.50-7.47 (m, 4H, Ar—H), 7.46-7.42 (m, 7H, Ar—H), 7.40-7.37 (m, 5H, Ar—H), 7.36-7.32 (m, 5H, Ar—H), 7.30-7.27 (m, 10H, Ar—H), 7.27-7.24 (m, 7H, Ar—H), 7.24-7.21 (m, 5H, Ar—H), 7.21-7.19 (m, 3H, Ar—H), 7.19-7.16 (m, 3H, Ar—H), 5.64 (s, 1H), 5.59 (s, 1H), 5.52 (d, J=3.1 Hz, 1H), 5.49 (d, J=2.8 Hz, 1H), 5.30-5.24 (m, 3H), 5.19-5.09 (m, 4H), 5.05-4.96 (m, 5H), 4.89-4.86 (m, 2H), 4.81-4.78 (m, 2H), 4.68-4.62 (m, 6H, ArCH2), 4.60-4.54 (m, 7H), 4.53-4.49 (m, 4H), 4.41-4.36 (m, 4H), 4.33-4.27 (m, 3H), 4.25-4.18 (m, 6H), 4.16-4.12 (m, 3H), 4.10-4.04 (m, 3H), 4.03-3.98 (m, 3H), 3.97-3.93 (m, 3H), 3.92-3.89 (m, 2H), 3.88-3.83 (m, 3H), 3.69 (t, J=9.4 Hz, 1H), 3.60-3.49 (m, 2H), 3.40-3.36 (m, 1H), 3.29-3.18 (m, 3H), 3.13 (d, J=9.1 Hz, 1H), 1.60-1.47 (m, 4H, CH2 in linker), 1.40-1.28 (m, 2H, CH2 in linker); 13C NMR (150 MHz, CD3OD): δ 176.4 (C), 176.1 (C), 175.9 (C), 158.6/158.0 (C), 140.0 (C), 139.9 (C), 139.5 (C), 139.4 (C), 139.3 (C), 139.2 (C), 139.14 (C), 139.06 (C), 139.0 (C), 138.2 (C), 138.1 (C), 137.2 (C), 134.8 (C), 134.5 (C), 132.4 (CH), 132.3 (CH), 132.2 (CH), 131.7 (CH), 131.0 (CH), 130.4 (CH), 129.8 (CH), 129.72 (CH), 129.67 (CH), 129.6 (CH), 129.5 (CH), 129.44 (CH), 129.39 (CH), 129.3 (CH), 129.23 (CH), 129.17 (CH), 129.1 (CH), 129.04 (CH), 128.99 (CH), 128.92 (CH), 128.86 (CH), 128.8 (CH), 128.7 (CH), 128.5 (CH), 128.4 (CH), 128.3 (CH), 128.1 (CH), 127.6 (CH), 127.1 (CH), 126.9 (CH), 122.2 (C), 122.09 (C), 122.07 (C), 102.8 (CH), 102.0 (CH), 98.3 (C), 98.0 (CH), 97.1 (CH), 96.9 (CH), 96.5 (CH), 85.2 (CH), 85.0 (CH), 82.0 (CH), 81.8 (CH), 81.4 (CH), 80.3 (CH), 79.3 (CH), 78.9 (CH), 78.6 (CH), 78.5 (CH), 78.3 (CH), 78.1 (CH), 76.9 (CH), 76.1 (CH2), 75.9 (CH), 75.5 (CH2), 75.3 (CH2), 73.2 (CH2/CH), 73.0 (CH2/CH), 72.6 (CH), 72.4 (CH), 71.65 (CH), 71.56 (CH), 71.2 (CH2), 71.0 (CH), 70.1 (CH), 69.31 (CH), 69.27 (CH), 68.9 (CH2), 68.5 (CH2), 68.4 (CH2), 67.5 (CH2), 65.7 (CH), 65.5 (CH), 65.2 (CH), 65.1 (CH), 51.7 (CH2), 51.4 (CH2), 50.0 (CH2), 47.8 (CH2), 30.2 (CH2), 29.2 (CH2), 28.6 (CH2), 24.2 (CH2); HRMS (MALDI): m/z calcd for C135H133Br4N13O67S8Na14 ([M+14Na−12H]2+): 1953.7903, found: 1953.7806.
  • (2R,3S,4S,5R,6R)-3-(((2R,3R,4R,5S,6R)-3-amino-4,5-dihydroxy-6-((sulfon atooxy)methyl)tetrahydro-2H-pyran-2-yl)oxy)-6-(((2R,3S,4R,5R,6R)-5-amino-6-(((2S,3S,4S,5R,6R)-6-(((2R,3S,4R,5R,6R)-5-amino-6-(((2R,3S,4S,5R,6R)-6-(((2R,3S,4R,5R,6R)-5-amino-6-(((2S,3S,4S,5R,6R)-6-((5-aminopentyl)oxy)-2-carboxylato-4-hydro xy-5-(sulfonatooxy)tetrahydro-2H-pyran-3-yl)oxy)-4-hydroxy-2-((sulfonatooxy)me thyl)tetrahydro-2H-pyran-3-yl)oxy)-2-carboxylato-4-hydroxy-5-(sulfonatooxy)tetr ahydro-2H-pyran-3-yl)oxy)-4-hydroxy-2-((sulfonatooxy)methyl)tetrahydro-2H-pyr an-3-yl)oxy)-2-carboxylato-4-hydroxy-5-(sulfonatooxy)tetrahydro-2H-pyran-3-yl)o xy)-4-hydroxy-2-((sulfonatooxy)methyl)tetrahydro-2H-pyran-3-yl)oxy)-4-hydroxy-5-(sulfonatooxy)tetrahydro-2H-pyran-2-carboxylate (19)
  • A solution of the compound 18 and Pd(OH)2 on carbon (3 g/g of the octasaccharide) in phosphate buffer (pH=7.0, 15 mL/g of the octasaccharide) with added MeOH as necessary to improve solubility of the octasaccharide was equipped with a hydrogen balloon and the mixture was stirred at room temperature for 2 d. The whole mixture was filtered through Celite and the filtrate was concentrated in vacuo. The residue was purified through a Sephadex G-25 column with H2O as eluent followed by Na+ exchange through AG 50WX8 (Na+ form) cation exchange resin. Lyophilization of the appropriate fractions afforded the desired product 19 (80%).
  • 1H NMR (600 MHz, D2O) δ 5.52 (d, J=3.8 Hz, 1H), 5.46 (d, J=3.7 Hz, 1H), 5.31 (t, J=4.4 Hz, 1H), 5.16 (d, J=4.1 Hz, 1H), 4.84 (d, J=1.8 Hz, 1H), 4.80 (brs, 1H), 4.52 (d, J=7.8 Hz, 1H), 4.47 (d, J=11.0 Hz, 1H), 4.29-4.25 (m, 4H), 4.19-4.07 (m, 7H), 4.01 (dd, J=8.8, 7.9 Hz, 1H), 3.97-3.91 (m, 2H), 3.90-3.70 (m, 14H), 3.64 (t, J=9.8 Hz, 1H), 3.62-3.59 (m, 1H), 3.47 (t, J=9.7 Hz, 1H), 3.28 (dd, J=10.7, 3.8 Hz, 1H), 3.23-3.16 (m, 2H), 2.91 (t, J=7.3 Hz, 2H), 1.63-1.53 (m, 4H), 1.44-1.36 (m, 2H).; 13C NMR (150 MHz, D2O) δ 175.3, 175.0, 174.6, 174.6, 100.5, 100.0, 98.8, 98.5, 96.4, 96.0, 91.7, 91.4, 80.0, 79.6, 77.1, 76.9, 76.1, 76.0, 75.8, 75.1, 75.0, 74.9, 72.9, 72.7, 70.8, 70.4, 70.2, 70.1, 69.7, 69.5, 69.3, 68.9, 68.6, 68.4, 67.7, 67.2, 67.1, 66.1, 66.0, 65.4, 63.1, 62.8, 54.3, 54.1, 53.9, 39.4, 27.9, 26.1, 21.8; HRMS (ESI): m/z calcd for C53H87N5Na4O65S8 ([M+10H−8Na]2+): 1090.5501, found: 1090.5498.
  • Compound 20:
  • To the octasaccharide amine compound 19 in 10% water in DMF (220 μL/1 mg), Et3N (25 μL/1 mg) and freshly prepared activated biotin ester (75 μL/1 mg of 0.1M in DMF, 15 eq) were added at rt. Stirring was continued for another 16 h at rt. Evaporated the solvent using N2 gas stream and purified using G-10 sephadex size exclusive column chromatography followed by Na+ exchange by passing through AG 50WX8 (Na+ form) cation exchange resin column. Lyophilization of the appropriate fractions afforded the desired compound 20 as a white foam solid in 94%.
  • 1H NMR (600 MHz, D2O) δ 5.37-5.26 (m, 1H), 5.10 (s, 1H), 5.08-5.04 (m, 1H), 4.62-4.60 (m, 1H), 4.54-4.50 (m, 3H), 4.50-4.41 (m, 2H), 4.35-4.32 (m, 3H), 4.30-4.20 (m, 4H), 4.13 (brs, 3H), 4.06-4.00 (m, 1H), 3.97 (brs, 2H), 3.93-3.88 (m, 4H), 3.83-3.76 (m, 2H), 3.76-3.63 (m, 21H), 3.70-3.53 (m, 120H), 3.59-3.51 (m, 20H), 3.52-3.43 (m, 1H), 3.31-3.29 (m, 10H), 3.26-3.23 (m, 6H), 3.09 (t, J=6.8 Hz, 1H), 3.02 (d, J=13.3 Hz, 1H), 2.95-2.85 (m, 4H), 2.83 (dd, J=13.3, 4.7 Hz, 1H), 2.70 (s, 2H), 2.68 (s, 1H), 2.62-2.54 (m, 2H), 2.52-2.50 (m, 2H), 2.42 (t, J=6.2 Hz, 1H), 2.38-2.36 (m, 4H), 2.20-2.17 (m, 10H), 1.68-1.40 (m, 21H), 1.34-1.31 (m, 10H); 13C NMR (150 MHz, D2O) δ 180.2, 180.1, 176.9, 176.9, 175.2, 174.6, 174.4, 174.2, 174.0, 173.7, 165.3, 165.3, 163.1, 160.2, 100.4, 99.9, 99.1, 98.9, 97.1, 96.9, 94.4, 94.0, 80.3, 79.9, 76.5, 76.2, 75.5, 74.2, 71.6, 71.0, 70.4, 69.6, 69.6, 69.5, 69.5, 69.4, 69.4, 69.3, 69.3, 68.8, 67.9, 66.8, 66.7, 66.7, 66.5, 66.4, 66.2, 65.0, 64.4, 62.1, 60.2, 59.5, 59.5, 58.4, 55.3, 55.3, 55.1, 53.6, 53.2, 52.9, 50.2, 40.9, 39.7, 39.7, 39.4, 38.9, 38.9, 37.6, 37.1, 36.1, 35.9, 35.8, 35.7, 35.6, 35.4, 35.4, 28.5, 28.3, 27.9, 27.8, 27.7, 27.7, 27.6, 25.1, 25.1, 22.4; HRMS (ESI): m/z calcd for C178H299N20O110S13 ([M+7H−12Na]5−): 979.0973, found: 979.0985.
  • Example 2 CTC Detection by Use of Magnetic Beads Pre-Coated with the Compound of Example 1
  • A microfluid chip for capturing CTCs from 65 advanced or metastatic CCA samples was constructed, and CTCs in all 65 bold samples were respectively captured using magnetic beads pre-coated with the compound of Example 1 (i.e., MB-SCH45) or an EpCAM antibody (i.e., MB-anti-EpCAM, as a control) in accordance with the procedures described in the “Materials and Methods” section.
  • It was found that MB-SCH45 was capable of isolating CTCs from all samples, regardless of the disease status. The number of CTCs isolated from the above samples using MB-SCH45 and MB-anti-EpCAM and details regarding the stage and organ of metastasis have been presented in Table 1. It is noteworthy that CTCs could be detected even in the blood of patients (Nos. 5, 14, 23, 30, 32, 43, and 60 in Table 1) with no distant metastasis as observed by radiographic imaging at the time of observation. We also observed that the size of the CTCs ranged from 5 to 20 μm in diameter (data not shown). Most of the cells presented a high nucleus-to-cytoplasmic ratio but not in all cases. We also documented dramatic heterogeneity in terms of cell shapes among patients as has been documented in other studies. Experimental data revealed that MB-SCH45 and MB-anti-EpCAM led to the isolation of ≥1 and ≥3 CTCs mL−1 of blood respectively. Furthermore, several stage-IV patients being characterized by the lowest concentrations and nonmetastatic patients displaying significant number of CTCs was also observed.
  • TABLE 1
    Number of CTCs isolated/mL of blood. Information of the disease, organ of metastasis,
    stage and number of CTCs isolated using MB-SCH45 or MB-anti-EpCAM
    No. of CTC isolated
    Sample Organ of MB- MB-anti-
    # Disease metastasis Stage SCH45 EpCAM
    1 Intrahepatic Distant IV 12 28
    cholangiocarcinoma lymph nodes
    2 Intrahepatic Peritoneal IV 3 10
    cholangiocarcinoma seeding
    3 Hilar Neck lymph IV 4 20
    cholangiocarcinoma nodes
    4 Intrahepatic lymph nodes IV 45 122
    cholangiocarcinoma metastasis
    and peritoneal
    seeding
    5 Intrahepatic No distant III 24 90
    cholangiocarcinoma metastasis
    6 Intrahepatic Lymph nodes III 30 98
    cholangiocarcinoma
    7 Intrahepatic Peritoneal IV 3 10
    cholangiocarcinoma seeding
    8 Intrahepatic Brain IV 6 15
    cholangiocarcinoma metastasis
    9 Hilar Neck lymph IV 6 20
    cholangiocarcinoma nodes
    10 Intrahepatic Liver IV 38 126
    cholangiocarcinoma metastasis
    11 Intrahepatic lymph nodes IV 50 121
    cholangiocarcinoma metastasis
    and peritoneal
    seeding
    12 Intrahepatic Lymph nodes IV 15 45
    cholangiocarcinoma
    13 Intrahepatic Lymph nodes III 4 15
    cholangiocarcinoma
    14 Intrahepatic No distant III 12 15
    cholangiocarcinoma metastasis
    15 Intrahepatic Lymph nodes III 60 120
    cholangiocarcinoma
    16 Hilar Adrenal IV 70 104
    cholangiocarcinoma gland
    17 Intrahepatic Peritoneal IV 47 102
    cholangiocarcinoma seeding
    18 Intrahepatic Peritoneal IV 84 126
    cholangiocarcinoma seeding
    19 Intrahepatic Lymph nodes III 47 123
    cholangiocarcinoma
    20 Intrahepatic Lymph nodes III 9 30
    cholangiocarcinoma
    21 Hilar Neck lymph IV 7 30
    cholangiocarcinoma nodes
    22 Intrahepatic Lymph nodes IV 6 20
    cholangiocarcinoma
    23 Intrahepatic No distant III 5 20
    cholangiocarcinoma metastasis
    24 Intrahepatic Distant IV 3 10
    cholangiocarcinoma lymph nodes
    25 Intrahepatic Lymph node III 30 50
    cholangiocarcinoma recurrence
    26 Intrahepatic Peritoneal IV 38 75
    cholangiocarcinoma seeding
    27 Intrahepatic Lymph nodes III 38 121
    cholangiocarcinoma
    28 Intrahepatic Liver IV 35 80
    cholangiocarcinoma metastasis
    29 Hilar lymph nodes III 16 50
    cholangiocarcinoma
    30 Intrahepatic No distant III 3 20
    cholangiocarcinoma metastasis
    31 cholangiocarcinoma Lymph nodes IV 3 7
    32 Intrahepatic No distant IV 7 25
    cholangiocarcinoma metastasis
    33 Intrahepatic liver IV 13 25
    cholangiocarcinoma
    34 Intrahepatic Peritoneal IV 17 30
    cholangiocarcinoma seeding
    35 cholangiocarcinoma Lymph nodes III 1 5
    36 cholangiocarcinoma Lymph nodes IV 30 50
    37 cholangiocarcinoma Lymph nodes III 19 35
    38 cholangiocarcinoma Lymph nodes IV 10 25
    39 Intrahepatic Peritoneal IV 4 7
    cholangiocarcinoma seeding
    40 Intrahepatic liver IV 22 55
    cholangiocarcinoma
    41 cholangiocarcinoma Lymph nodes IV 1 3
    42 cholangiocarcinoma Lymph nodes IV 6 12
    43 Intrahepatic No distant III 2 5
    cholangiocarcinoma metastasis
    44 Intrahepatic Lymph nodes IV 3 8
    cholangiocarcinoma
    45 Intrahepatic No distant IV 6 16
    cholangiocarcinoma metastasis
    46 Intrahepatic liver IV 2 6
    cholangiocarcinoma
    47 Intrahepatic Peritoneal IV 27 64
    cholangiocarcinoma seeding
    48 cholangiocarcinoma Lymph nodes III 11 24
    49 Intrahepatic Lymph nodes IV 1 4
    cholangiocarcinoma
    50 cholangiocarcinoma Lymph nodes III 1 3
    51 Intrahepatic liver IV 55 98
    cholangiocarcinoma
    52 cholangiocarcinoma Lymph nodes IV 72 128
    53 Intrahepatic liver III 97 200
    cholangiocarcinoma
    54 Intrahepatic Lymph nodes IV 12 36
    cholangiocarcinoma
    55 Intrahepatic Lymph nodes IV 1 3
    cholangiocarcinoma
    56 Intrahepatic Lymph nodes IV 5 12
    cholangiocarcinoma
    57 cholangiocarcinoma Lymph nodes III 5 10
    58 Intrahepatic liver IV 9 22
    cholangiocarcinoma
    59 Intrahepatic liver IV 1 3
    cholangiocarcinoma
    60 Multiple Intrahepatic No distant IV 4 9
    cholangiocarcinoma metastasis
    61 Intrahepatic Lymph nodes IV 1 4
    cholangiocarcinoma
    62 Intrahepatic Lymph nodes IV 5 12
    cholangiocarcinoma
    63 cholangiocarcinoma Lymph nodes III 1 4
    64 Intrahepatic liver III 23 59
    cholangiocarcinoma
    65 Multiple Intrahepatic liver IV 15 37
    cholangiocarcinoma
  • The immunofluorescence images of some samples are shown in FIG. 2. It is worth mentioning that the samples were randomly selected and in no particular order. CTC clusters could be observed using both MB-SCH4 and MB-anti-EpCAM in some stage IV patients. Some clusters were relatively small while others comprised dozens of tumor cells. The CTC clusters were more evident when MB-anti-EpCAM was used for positive enrichment and have been shown in FIG. 2 (the bottom-most 4 panels). Such cluster-forming CTCs are known to have a relatively higher potential for metastasis and should be studied in detail in future works.
  • Example 3 CTCs in Assessing Disease Status
  • In this example, the correlation between CTC enumeration and treatment response based on standard radiographic examination was investigated by analyzing medical records of 5 patients with advanced or metastatic CCA (i.e., patients A to E). Detailed information of these 5 patients with treatment strategy and regiments is summarized in Table 2. The number of CTCs and tumor size before treatment were treated as 100%, and the after-treatment data pertaining to individual cell number profiles and tumor size measurements were normalized to the above. The results of normalized CTC isolation using MB-SCH45, MB-anti-EpCAM, and flow assisted cell sorting (FACS) alongside computed tomography (CT) scan tumor measurements are illustrated in FIG. 3. It was found that CTCs could be successfully detected in all patients, even after several lines of chemotherapy. The total number of CTCs after the first round of treatment decreased when compared with that before treatment in all cases and corresponded with the radiographic response in all cases tested. An increased number of CTCs was observed in patients A, C and D after few lines of chemotherapy which later seemed to corroborate the radiographic finding that the tumor size had increased in these patients. These results not only implied the predictive role of CTCs for therapeutic efficacies in all patients, but also appeared to prognosticate the response to chemotherapy, often predicting the efficacy of treatment before radiographic results. The data were comparable using MB-SCH45 (patients A, B and D), or even better (patients C and E) than FACS analysis. Similar trend was also observed using MB-EpCAM. Taken together, the results demonstrated a prognostic and predictive relevance of persisting CTCs during chemotherapy, indicating a potential role of the present compound of formula (I) in monitoring of CCA treatment.
  • TABLE 2
    Information of patients A to E.
    Type of
    Patient/Age/Sex Disease Stage chemotherapy
    Patient A/47/F Intrahepatic IV S-1a, leucovorin
    cholangiocarcinoma (LV), oxaliplatin
    with peritoneal and gemcitabine
    seeding (SLOG)
    Patient B/65/M cholangiocarcinoma IV gemcitabine and
    with lymph cisplatin
    node metastasis
    Patient C/83/F Intrahepatic III Concurrent
    cholangiocarcinoma radiotherapy
    with no distant followed by oral
    metastasis chemotherapy of
    S-1
    Patient D/46/M Multiple IV S-1, leucovorin
    Intrahepatic (LV), oxaliplatin
    cholangiocarcinoma and gemcitabine
    with vessel (SLOG)
    invasion
    Patient E/66/F Cholangiocarcinoma IV S-1, leucovorin
    with lymph (LV), oxaliplatin
    node metastasis and gemcitabine
    (SLOG)

    a: S-1 is a combination of tegafur, 5-chloro-2,4-dihydropyrimidine (CDHP), and potassium oxonate.
  • It will be understood that the above description of embodiments is given by way of example only and that various modifications may be made by those with ordinary skill in the art. The above specification, examples and data provide a complete description of the structure and use of exemplary embodiments of the invention. Although various embodiments of the invention have been described above with a certain degree of particularity, or with reference to one or more individual embodiments, those with ordinary skill in the art could make numerous alterations to the disclosed embodiments without departing from the spirit or scope of the present disclosure.

Claims (12)

What is claimed is:
1. A compound of formula (I),
Figure US20220373552A1-20221124-C00015
wherein,
R1 and R2 are independently H, or —SO3M; and M is a monovalent cation selected from the group consisting of sodium ion, potassium ion, lithium ion or ammonium ion.
2. The compound of claim 1, wherein in the formula (I), R1 and R2 are independently —SO3M, and M is the sodium ion.
3. A method of treating and detecting circulating cholangio-cancerous cells from a biological sample of a subject suspected of having cholangiocarcinoma (CCA) comprising:
(a) contacting the biological sample with the compound of claim 1; and
(b) detecting a complex formed between the compound of claim 1 and the biological sample in an immunoassay; and
(c) administering to the subject an effective amount of a chemotherapeutic agent to ameliorate symptoms associated with the CCA.
4. The method of claim 3, wherein in the formula (I), R1 and R2 are independently —SO3M, and M is the sodium ion.
5. The method of claim 3, wherein the compound of claim 1 is coupled to streptavidin, which is pre-conjugated on the outer surface of a magnetic bead.
6. The method of claim 3, wherein the biological sample is selected from the group consisting of blood, plasma, serum, urine, sputum, saliva, tissue sample, biopsy, and tissue lysate.
7. The method of claim 6, wherein the biological sample is the blood.
8. The method of claim 7, further comprising pre-treating the blood with a lysis buffer to lyse red blood cells therein before the step (a).
9. The method of claim 3, wherein the subject has stage III or IV CCA, or metastatic CCA.
10. The method of claim 3, wherein in the step (c), the chemotherapeutic agent is selected from the group consisting of S-1, leucovorin, oxaliplatin, gemcitabine, cisplatin, and a combination thereof.
11. The method of claim 10, the S-1 is a combination of tegafur, 5-chloro-2,4-dihydropyrimidine (CDHP), and potassium oxonate.
12. The method of claim 10, wherein in the step (c), the therapeutic agent is a combination of gemcitabine and cisplatin.
US17/238,431 2021-04-23 2021-04-23 Methods for detecting and treating cholangiocarcinoma Active 2041-07-01 US11506664B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/238,431 US11506664B1 (en) 2021-04-23 2021-04-23 Methods for detecting and treating cholangiocarcinoma

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US17/238,431 US11506664B1 (en) 2021-04-23 2021-04-23 Methods for detecting and treating cholangiocarcinoma

Publications (2)

Publication Number Publication Date
US11506664B1 US11506664B1 (en) 2022-11-22
US20220373552A1 true US20220373552A1 (en) 2022-11-24

Family

ID=84103658

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/238,431 Active 2041-07-01 US11506664B1 (en) 2021-04-23 2021-04-23 Methods for detecting and treating cholangiocarcinoma

Country Status (1)

Country Link
US (1) US11506664B1 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI20011671A (en) * 2001-08-20 2003-02-21 Carbion Oy Tumor-specific oligosaccharide sequences and their use

Also Published As

Publication number Publication date
US11506664B1 (en) 2022-11-22

Similar Documents

Publication Publication Date Title
US10935544B2 (en) Glycan arrays and method of use
US10662212B2 (en) Carbohydrate ligands that bind to IGM antibodies against myelin-associated glycoprotein
Alon‐Maimon et al. Fusobacterium nucleatum and cancer
JP2013049718A (en) Glycomimetic inhibitor of pa-il lectin, pa-iil lectin or both the lectins from pseudomonas
TW201808978A (en) Methods for modular synthesis of N-glycans and arrays thereof
JP2014532049A (en) Pharmaceutical compounds for use in the treatment of Clostridium difficile infection
US20200323893A1 (en) Detection of metastatic disease and related methods
US11506664B1 (en) Methods for detecting and treating cholangiocarcinoma
BR112019019145A2 (en) glycopolymer sequestering carbohydrate-binding proteins
Subramaniam et al. Anti-α-Gal antibodies detected by novel neoglycoproteins as a diagnostic tool for Old World cutaneous leishmaniasis caused by Leishmania major
Di Leo et al. Predictive molecular markers in the adjuvant therapy of breast cancer: state of the art in the year 2002
CN108367059A (en) For the synthetic vaccine of S. pneumoniae serotypes 2
TWI778596B (en) Compounds and methods for detecting cholangio-cancerous cells
US20210046479A1 (en) Devices and methods for detecting cancerous cells
US20220305001A1 (en) Methods of treating acute myeloid leukemia with farnesyltransferase inhibitors
WO2014170681A9 (en) Oligosaccharide conjugates and methods of use
JP6595460B2 (en) Glycoclusters and their use as antibacterial agents
US10806730B2 (en) Methods of treating cancer with farnesyltransferase inhibitors
Seki et al. Synthesis and Immunological Evaluation of Escherichia coli O1-Derived Oligosaccharide–Protein Conjugates toward Avian Pathogenic Escherichia coli O1 Vaccine Development
TW202313062A (en) Synthetic compound, kit comprising the same, and uses thereof
US20210330693A1 (en) Heparanase inhibitors and their use as anti-cancer compounds
WO2023070121A1 (en) Compositions and methods for treatment of mic60 depleted cancers and metastasis
JPH1081627A (en) Blood coagulation promoter
WO2022038196A1 (en) Method and kit for labeling eukaryotic cells from a multicellular organism using modified monosaccharide compounds and pharmaceutical composition comprising such cells
CN117642168A (en) Inhibitors of bruton's tyrosine kinase and methods of use thereof

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE