[go: up one dir, main page]

WO2025176152A1 - Probiotics mixtures and methods of use thereof for treating cancers - Google Patents

Probiotics mixtures and methods of use thereof for treating cancers

Info

Publication number
WO2025176152A1
WO2025176152A1 PCT/CN2025/078098 CN2025078098W WO2025176152A1 WO 2025176152 A1 WO2025176152 A1 WO 2025176152A1 CN 2025078098 W CN2025078098 W CN 2025078098W WO 2025176152 A1 WO2025176152 A1 WO 2025176152A1
Authority
WO
WIPO (PCT)
Prior art keywords
prohep
composition
cancer
hcc
crc
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2025/078098
Other languages
French (fr)
Inventor
Hani EI-NEZAMI
Fangfei ZHANG
Kwun Kwan LO
Hoi Kit Matthew LEUNG
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.)
University of Hong Kong HKU
Original Assignee
University of Hong Kong HKU
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 University of Hong Kong HKU filed Critical University of Hong Kong HKU
Publication of WO2025176152A1 publication Critical patent/WO2025176152A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/745Bifidobacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the disclosed invention is generally in the field of cancer treatment and specifically in the area of treating cancers with probiotics.
  • Metabolic dysfunction-associated steatotic liver disease is a prevalent and multifaceted chronic hepatic disorder. It encompasses a range of pathological stages, including steatosis, metabolic dysfunction-associated steatohepatitis (MASH) , fibrosis, cirrhosis.
  • MASH metabolic dysfunction-associated steatohepatitis
  • MASLD elevates the risk of cardiovascular events, chronic kidney disease, as well as hepatic and extrahepatic malignancies, notably hepatocellular carcinoma (HCC) .
  • HCC hepatocellular carcinoma
  • the incidence of MASLD and its associated complications, particularly HCC is anticipated to escalate in the foreseeable future. HCC is the second leading cause of cancer-related mortality globally. Nonetheless, no efficacious therapeutic interventions currently exist for MASLD-HCC.
  • the portal venous system supplies over two-thirds of the liver's blood from the intestinal tract, allowing intestinal bacteria and their constituents to efficiently reach the hepatic system.
  • the translocated elements such as bacterial products and lipopolysaccharides, can stimulate hepatic immune cells, activate inflammatory pathways, and ultimately contribute to the development of MASLD-HCC.
  • probiotics The manipulation of gut microbiota using probiotics may offer a promising approach for liver diseases.
  • Animal studies have demonstrated that probiotics can confer beneficial effects against MASLD.
  • numerous studies have reported the efficacy of probiotics as a prophylactic against different cancer types and as an adjuvant for cancer therapy to enhance its effectiveness.
  • most studies have primarily investigated the early stages of MASLD, including liver steatosis and inflammation, while neglecting liver fibrosis, which is a hallmark of the advanced phase.
  • probiotics in MASLD-HCC remains poorly understood.
  • Commensal probiotics from some genera such as lactobacillus, bifidobacterium and Butyricimonas in the gut were found to be diminished in MASLD-HCC patients.
  • CRC Colorectal cancer
  • Surgical treatments including endoscopic mucosal resection and endoscopic submucosal dissection in the early stages and lymph node dissection in the advanced stages along with polyps removal, are the standard first-line treatment for CRC.
  • chemotherapeutic intervention is crucial to prevent and cure metastatic CRC and improve patients’ s urvival rates. Nonetheless, the response rate of the standard chemo drug backbone 5-fluorouracil (5-FU) on CRC is as low as 11%without combining with other regimens. Given the high economic burden of chemotherapy, affordable and effective pharmaceutical options are in demand to save lives.
  • Gut microbiota and cancer exhibit a bidirectional relationship, in which the alteration of microbial composition could promote pathogenesis while the modulation of gut microbiota could also be therapeutic for cancer.
  • Gut microbiota’s influence on CRC initiation and progression was suggested to be both the “driver” and the “passenger” of the “driver/passenger” theory.
  • the imbalance of the gut microbiota profile termed gut dysbiosis was found to be a vital cause of colorectal tumorigenesis.
  • modulation of the gut microbiota alleviated or prevented the progression of CRC in both healthy and cancerous subjects.
  • Microbiome-targeted therapy in CRC treatment includes fecal microbiota transplant (FMT) , probiotics, diet and prebiotics, and antibiotic treatment. Unlike the complexity and high cost of personalized FMT, the damaging effects of antibiotics to commensal bacteria, the induction of probiotics in CRC treatment is a more affordable and efficient option for modulating the gut microbial composition.
  • FMT fecal microbiota transplant
  • probiotics Unlike the complexity and high cost of personalized FMT, the damaging effects of antibiotics to commensal bacteria, the induction of probiotics in CRC treatment is a more affordable and efficient option for modulating the gut microbial composition.
  • CRC colorectal cancer
  • 5-FU is an essential component in the standard chemotherapy of CRC and yet its efficacy was limited due to the development of chemoresistance and toxicity.
  • Adjuvant combinations of probiotics with 5-FU were found to improve 5-FU applicability in CRC management.
  • the anti-cancer effects of 5-FU were enhanced with reduced intestinal toxicity and chemoresistance effects.
  • compositions and methods for treating and/or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma using a probiotic formulation, Prohep. Also described are methods for treating and/or preventing cancer, particularly hepatocellular carcinoma and colorectal cancer using the probiotic formulation.
  • the probiotic formulation, Prohep includes a mix of organisms selected from a group that includes Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus GG, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium animalis subsp. lactis and Streptococcus thermophilus.
  • Prohep is formulated for oral administration.
  • Prohep compositions may also be administered orally, intracolonically, intranasally, intrarectally, via a catheter, via a lavage, via a nasogastric tube, via local delivery, or via a method for fecal microbiota transplantation (FMT) .
  • Prohep composition may be in the form of a dispersion. Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils.
  • the current disclosure uses Prohep as an adjuvant therapy in combination with low-dose Sorafenib for the treatment of MASLD-HCC.
  • the Prohep can be used alone for the treatment of MASLD-HCC.
  • Prohep exhibits anti-tumorigenic properties by modulating the gut microbiota to increase the production of short-chain fatty acids (SCFAs) , including propionate, valerate, and butyrate, thereby improving gut dysbiosis and enhancing treatment efficacy.
  • SCFAs short-chain fatty acids
  • the activation of AMPK by Prohep results in reduced lipogenesis, decreased lipid uptake, and upregulation of antioxidant enzyme expressions.
  • Prohep suppresses the PI3K/mTOR cancer proliferation pathway, contributing to reduced tumor counts, amelioration of inflammation, and increased hepatic superoxide dismutase (SOD) expression.
  • SOD superoxide dismutase
  • the combination of Prohep and low-dose Sorafenib leads to improved therapeutic outcomes without inducing adverse effects, as evidenced by decreased proliferation marker Ki67 expression, alleviation of hepatic inflammation, and maintenance of redox balance.
  • the current disclosure highlights the modulation of Sorafenib-treated gut microbiota by Prohep, resulting in enhanced butyrate production and increased sensitivity to Sorafenib, establishing an important therapeutic approach for the neoadjuvant and long-term management of MASLD-HCC.
  • the present disclosure uses Prohep for treating colorectal cancer.
  • Prohep demonstrates enhanced anti-tumorigenic effects compared to 5-Fluorouracil (5-FU) alone or in combination for treating colorectal cancer.
  • Prohep alleviates AOM/DSS-induced colorectal tumorigenesis by modulating inflammatory, proliferative, and apoptotic pathways, including downregulation of TNF- ⁇ and p-STAT3 and upregulation of p53.
  • Prohep reduces tumor burden, caecum weight, crypt depth, colonic inflammation, and collagen fibrosis induced by AOM/DSS.
  • Prohep enriches beneficial bacteria such as Helicobacter ganmani, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila, Desulfovibrio porci, and Prevotella sp. PTAC, which were inversely correlated with tumor counts and suppressed AOM/DSS-associated pathogenic bacteria.
  • Prohep downregulated pathways associated with colorectal cancer (CRC) including those involved in peptidoglycan, lipopolysaccharide (LPS) , and uric acid biosynthesis and conversion.
  • Prohep enhanced pathways related to CRC-suppressing metabolites such as menaquinone, tetrapyrrole, aminolevulinic acid, and tetrahydrofolate.
  • Prohep enhanced the biosynthesis of beneficial compounds like L-lysine, lipoic acid, pyrimidine, and palmitate and promoted metabolic pathways supporting energy utilization by lactic acid-producing bacteria (LAB) and acetate producers.
  • LAB lactic acid-producing bacteria
  • Prohep further upregulated fecal acetate concentration, contributing to its anti-CRC effects.
  • Figures 1A-1B show Prohep protects against NAFLD-HCC formation in DEN-HFD MASLD-HCC mouse models.
  • FIG. 1A Experimental design for the dietary plan and treatment scheme in DEN+HFHC mouse model.
  • FIG. 1B Representative gross morphology, H&E-stained tumor area (T circled with red dotted line indicates tumor area) (100x) , IHC staining (200x) for Ki-67 and C-Casp3 in liver and H&E-stained (40x) histological images in lung (arrow indicates lung metastasis) of DEN+HFHC mouse model.
  • Figure 2 shows Prohep ameliorated MASLD progression in DEN-HFD HCC mice model.
  • H&E (200x) Oil-red O (200x) , Sirius red-stained (200x) non-tumor area and IHC staining (200x) for F4/80.
  • Figures 3A-3B show that Prohep modulated tumor microenvironment via activating AMPK signaling pathway its impact on cancer metabolism.
  • FIG. 3A Representative IHC staining for AMPK images and scoring of liver tissues.
  • FIG. 3B Protein expression of FAS, HO-1 and FOXO3A. Data are presented as the mean ⁇ SD. *p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001, ****p ⁇ 0.0001.
  • Figure 4 shows that Prohep suppressed the activity of the PI3K/mTOR pathway in DEN-HFD induced HCC mouse models. Protein expression of P-PI3k, PI3K and mTOR. Data are presented as the mean ⁇ SD. *p ⁇ 0.05.
  • Figures 5A-5D show that Prohep altered gut microbiota–related metabolites.
  • Fig. 5A Significantly enriched KEGG pathways generated from fecal metagenomic data in Con and Pro groups.
  • Fig. 5B Significantly altered enzymes participates the pathway “Pyruvate fermentation to propanoate I” .
  • Fig. 5C Biosynthesis route of propionate.
  • Fig. 5D Fecal SCFAs measurements.
  • Figures 6A-6F shows Prohep enhanced survivability and reduced colorectal tumorigenesis of AOM/DSS mice better than 5-FU and Prohep+5-FU.
  • FIG. 6A Survival rate percentage.
  • FIG. 6B Representative macroscopic pictures of the dissected colon.
  • Fig. 6C Total tumor counts.
  • Fig. 6D Total tumor size.
  • Fig. 6E Caecum weight.
  • Fig. 6F Liver weight.
  • n 6-10. *p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001; ****p ⁇ 0.0001.
  • Figures 7A-7F show that Prohep improved colorectal histopathology induced by AOM/DSS.
  • FIG. 7A Colonic crypt depth.
  • FIG. 7B Hyperplasia score.
  • Fig. 7C Inflammation score.
  • Fig. 7D Sirius red positive percentage.
  • FIG. 7E Representative microscopic pictures of colonic sections under H&E staining. Colonic tumors were framed in rectangle and displayed in 100x.
  • FIGS 8A-8I Prohep suppressed pro-inflammatory and proliferative marker and promoted apoptotic markers.
  • FIG. 8C c-jun;
  • Fig. 8D c-fos;
  • Fig. 8E p-STAT3; and
  • Fig. 8F c-casp3.
  • FIG. 8H Representative immunoblots of p53.
  • FIG. 9A-9C Prohep modulated gut microbiota contributed to alleviate colorectal tumorigenesis and elevated SCFAs concentration.
  • FIG. 9A-9B Heatmap showing the z-score transformed mean abundance of significantly altered species in H, F, P or PF as compared to AD (Fig. 9A) and Spearman’s correlation between relative bacterial abundance and fecal SCFA concentrations, colonic cytokines concentration, tumor counts and colorectal tumorigenesis related organs alterations (Fig. 9B) .
  • FIG. 9C Fecal concentration of SCFA.
  • AA acetate
  • PA propionate
  • BA butyrate
  • IBA isobutyrate
  • VA valerate.
  • FIG. 10 Prohep enriched bacteria negatively correlated with AOM/DSS enriched bacteria. Inner correlation of enriched bacteria in AOM/DSS and Prohep group. Correlations with p-value ⁇ 0.05 and r > 0.3 were considered and visualized in the network. The green edge denotes for negative correlation and red edge denotes for positive correlation. The size of the node is proportional to the number of connections to other nodes.
  • FIG. 11A-11B Prohep modulated gut microbiota to induce anti-tumor metabolic functions.
  • Fig. 11A Heatmap showing the z-score transformed mean abundance of KEGG orthology (KO) originated from three significantly altered KEGG modules in H, F, P or PF as compared to AD (left) . Differential KO based on ANCOM-BC (adjusted p-value ⁇ 0.1) were displayed in the right part; and
  • Fig. 11B Significantly altered Metacyc pathway in H, F, T or TF as compared to AD.
  • the color is proportional to log2 transformed fold change (Treatment/Control) , while the size is proportional to the value of -log10 transformed adjusted p (FDR-corrected) .
  • n 6-10. *p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001; ****p ⁇ 0.0001.
  • Figure 12 Diagram summarizing the proposed mechanism of the disclosed probiotics mixture.
  • therapeutic agent refers to any agent that, when administered to a subject, has a therapeutic, prophylactic, and/or diagnostic effect and/or elicits a desired biological and/or pharmacological effect.
  • modulate refers to the ability of a compound to change an activity in some measurable way as compared to an appropriate control.
  • activities can increase or decrease as compared to controls in the absence of these compounds.
  • an increase in activity is at least 25%, more preferably at least 50%, most preferably at least 100%compared to the level of activity in the absence of the compound.
  • a decrease in activity is preferably at least 25%, more preferably at least 50%, most preferably at least 100%compared to the level of activity in the absence of the compound.
  • a compound that increases a known activity is an “agonist” .
  • One that decreases, or prevents, a known activity is an “antagonist” .
  • inhibitor means to reduce or decrease in activity or expression. This can be a complete inhibition or activity or expression, or a partial inhibition. Inhibition can be compared to a control or to a standard level. Inhibition can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98,
  • monitoring refers to any method in the art by which an activity can be measured.
  • providing refers to any means of adding a compound or molecule to something known in the art. Examples of providing can include the use of pipettes, pipettemen, syringes, needles, tubing, guns, etc. This can be manual or automated. It can include transfection by any mean or any other means of providing nucleic acids to dishes, cells, tissue, cell-free systems and can be in vitro or in vivo.
  • preventing refers to administering a compound prior to the onset of clinical symptoms of a disease or conditions so as to prevent a physical manifestation of aberrations associated with the disease or condition.
  • in need of treatment refers to a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals) that a subject requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a care giver's expertise, but that include the knowledge that the subject is ill, or will be ill, as the result of a condition that is treatable by the disclosed compounds.
  • a caregiver e.g. physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals
  • subject includes, but is not limited to, animals, plants, parasites and any other organism or entity.
  • the subject can be a vertebrate, more specifically a mammal (e.g., a human, horse, pig, rabbit, dog, sheep, goat, non-human primate, cow, cat, guinea pig or rodent) , a fish, a bird or a reptile or an amphibian.
  • the subject can be an invertebrate, more specifically an arthropod (e.g., insects and crustaceans) .
  • the term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered.
  • a patient refers to a subject afflicted with a disease or disorder.
  • patient includes human and veterinary subjects.
  • the subject can be any organism in which the disclosed method can be used to genetically modify the organism or cells of the organism.
  • the terms “individual” , “host” , “subject” , and “patient” are used interchangeably, and refer to a mammal, including, but not limited to, murids, simians, humans, mammalian farm animals and livestock, mammalian sport animals, and mammalian pets.
  • Treatment means to administer a composition to a subject or a system with an undesired condition (e.g., HCC) .
  • the condition can include one or more symptoms of a disease, pathological state, or disorder.
  • Treatment includes medical management of a subject with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder.
  • active treatment that is, treatment directed specifically toward the improvement of a disease, pathological state, or disorder
  • causal treatment that is, treatment directed toward removal of the cause of the associated disease, pathological state, or disorder.
  • this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological state, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological state, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological state, or disorder.
  • palliative treatment that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological state, or disorder
  • preventative treatment that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological state, or disorder
  • supportive treatment that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological state, or disorder.
  • Such measurements and assessments can be made in qualitative and/or quantitative terms.
  • characteristics or features of a disease, pathological condition, or disorder and/or symptoms of a disease, pathological condition, or disorder can be reduced to any effect or to any amount.
  • “Prevention” or “preventing” means to administer a composition to a subject or a system at risk for an undesired condition.
  • the condition can include one or more symptoms of a disease, pathological state, or disorder.
  • the condition can also be a predisposition to the disease, pathological state, or disorder.
  • the effect of the administration of the composition to the subject can be the cessation of a particular symptom of a condition, a reduction or prevention of the symptoms of a condition, a reduction in the severity of the condition, the complete ablation of the condition, a stabilization or delay of the development or progression of a particular event or characteristic, or reduction of the chances that a particular event or characteristic will occur.
  • the terms “effective amount” or “therapeutically effective amount” means a quantity sufficient to alleviate or ameliorate one or more symptoms of a disorder, disease, or condition being treated, or to otherwise provide a desired pharmacologic and/or physiological effect. Such amelioration only requires a reduction or alteration, not necessarily elimination. The precise quantity will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, weight, etc. ) , the disease or disorder being treated, as well as the route of administration, and the pharmacokinetics and pharmacodynamics of the agent being administered.
  • subject-dependent variables e.g., age, immune system health, weight, etc.
  • Microbial flora refers to the microorganisms that normally live in the gastrointestinal tract, skin, nose, etc.
  • the internal tissues e.g. blood, brain, muscle, etc.
  • the surface tissues i.e., skin and mucous membranes
  • Bacteria are the most numerous microbial components of the normal flora.
  • Microbiota a term created by Jeffrey Gordon, refers to the collection of microbial species that form a microbial community. This includes the normal flora and “harmful” ones.
  • Probiotic utilizes the World Health Organization's 2001 definition of “live micro-organisms which, when administered in adequate amounts, confer a health benefit on the host” . Probiotics must be alive when administered, have viability and reproducibility based on in vivo testing, and during use and storage.
  • drug regime refers to drug administration regarding formulation, route of administration, drug dose, dosing interval and treatment duration.
  • pharmaceutically acceptable refers to compositions, polymers and other materials and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to pharmaceutically acceptable materials, compositions or vehicles, such as a liquid or solid filler, diluent, solvent or encapsulating material involved in carrying or transporting any subject composition, from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be acceptable” in the sense of being compatible with the other ingredients of a subject composition and not injurious to the patient.
  • a “small molecule” is understood in the art to be an organic molecule that is less than about 2000 g/mol in size. In some forms, the small molecule is less than about 1500 g/mol or less than about 1000 g/mol. In some forms, the small molecule is less than about 800 g/mol or less than about 500 g/mol. In some forms, small molecules are non-polymeric and/or non-oligomeric. In some forms, small molecules are not proteins, peptides, or amino acids. In some forms, small molecules are not nucleic acids or nucleotides. In some forms, small molecules are not saccharides or polysaccharides.
  • the sub-group of A-E, B-F, and C-E are specifically contemplated and should be considered disclosed from disclosure of A, B, and C; D, E, and F; and the example combination A-D.
  • each of the materials, compositions, components, etc. contemplated and disclosed as above can also be specifically and independently included or excluded from any group, subgroup, list, set, etc. of such materials.
  • compositions and methods for treating and/or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma MASLD-HCC
  • colorectal cancer a metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma
  • Prohep for treating and/or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) and colorectal cancer.
  • MASLD-HCC metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma
  • Prohep The improved formulation of Prohep is listed as follow, Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium animalis subsp. lactis and Streptococcus thermophilus.
  • the old Prohep is composed of Lactobacillus rhamnosus GG (LGG) , viable Escherichia coli Nissle 1917 (EcN) and heat-inactivated VSL#3 (containing Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium infantis) .
  • the detailed components comparison was illustrated in Table 3.
  • the first main difference between the improved formula and old formula is that the following stains, Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium infantis are heat-inactivated in old formula, while all the stains are viable in the improved formula.
  • Cell viability has been recognized as a crucial factor for probiotics to provide health benefits. Having multiple probiotic strains that establish themselves in the gut can enhance gut microbiota diversity and promote a more resilient and balanced gut microbiota.
  • E. coli Nissle 1917 may provide certain health benefits, the taxonomic group it belongs, Escherichia coli is increasingly involved in various intestinal and extra-intestinal infections as an opportunistic pathogen.
  • Nonpathogenic and pathogenic strains of E. coli exhibit variations in their genetic makeup, which influence the development of specific virulence characteristics. But the genetic information responsible for these traits could be acquired through horizontal gene transfer and result in developing from commensal E. coli strains into pathogenic variants. Removal of E. coli Nissle 1917 diminishes the potential risk.
  • the third difference is the addition of Bifidobacterium animalis subsp. Lactis.
  • B. animalis subsp. Lactis is a renowned probiotic strain that have been proven its beneficial health effect and safety for consumption in numerous clinical studies. Clinical studies have confirmed its positive impact on gastrointestinal health and immune function by promoting a balanced gut microbiota, enhancing bowel function, mitigating antibiotic-related side effects, and more.
  • Anti-cancer agents or therapeutic agents include one or more of chemotherapeutics, targeted therapies, immunotherapies, hormonal therapies, epigenetic modulators, radiopharmaceuticals, angiogenesis inhibitors, natural products, nanoparticle-based therapies, RNA-based therapies, microbiome-modulating treatments, or a combination thereof.
  • anti-cancer agents that can be used in the pharmaceutical compositions containing the disclosed probiotic formulation or a separate pharmaceutical composition include, but are not limited to, temozolomide, carmustine, bevacizumab, procarbazine, lomustine, vincristine, gefitinib, erlotinib, cisplatin, carboplatin, oxaliplatin, 5-fluorouracil, gemcitabine, tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin, mithramycin, vinblastine, vindesine, vinorelbine, paclitaxel, taxol, docetaxel, etoposide, teniposide, amsacrine, topotecan, camptothe
  • the anti-cancer agent is Sorafenib.
  • Sorafenib brand name: Nexavar
  • Nexavar is an oral, multi-kinase inhibitor used to treat certain cancers, including hepatocellular carcinoma (liver cancer) , renal cell carcinoma (kidney cancer) , and radioactive iodine-resistant differentiated thyroid cancer. It works by blocking specific enzymes involved in tumor cell growth and blood vessel formation, targeting pathways like VEGFR, PDGFR, and RAF kinases. By inhibiting these pathways, sorafenib slows tumor progression and reduces blood supply to tumors.
  • Sorafenib is a multi-kinase inhibitor that is presently utilized as a first-line therapy for advanced HCC. Nevertheless, the limited response rate, rapid development of drug resistance, and severe and extensive adverse events associated with Sorafenib are major challenges to its clinical use. Adverse events have been associated with gut dysbiosis, which results in a less protective intestinal environment and increases the incident of diarrhea while probiotics supplementation could maintain a favorable gut microbiota against Sorafenib-induced disturbances. In clinical practice, the applied dose for HCC therapy is 800 mg/day.
  • Sorafenib (30 mg/kg) , which is equivalent to a human dose of 135 mg/day for a 60-kg person, was supplemented in mouse and monkey models at the initial stage of MASH-HCC.
  • the low dose of Sorafenib was found to effectively ameliorate major MASH hallmarks, including hepatic steatosis, inflammation, and fibrosis.
  • mice treated with low-dose Sorafenib experienced slight degrees of unrecovered hair, watery stool, and diarrhea. Additionally, declining systemic drug levels were observed in the mice, indicating the onset of Sorafenib resistance.
  • dosage of sorafenib can range from 30 mg/kg –800 mg/kg body weight, 25 mg/kg –700 mg/kg body weight, 20 mg/kg –600 mg/kg body weight, 15 mg/kg –500 mg/kg body weight, 10 mg/kg –400 mg/kg body weight, 5 mg/kg –300 mg/kg body weight, 1 mg/kg –200 mg/kg body weight, preferably 30 mg/kg body weight. In some forms, the dosage of sorafenib is less than 30 mg/kg.
  • Therapeutic agents for use in the disclosed methods for treatment of the disclosed subjects are provided.
  • the therapeutic agents are typically administered to a subject in an effective amount to treat the disease or disorder of the subject.
  • the therapeutic agent can be in a pharmaceutical composition.
  • the therapeutic agent is most typically a compound that reduces the biological activity of a target molecule.
  • the compound is an inhibitory polypeptide such as, but not limited to, an antibody; a small molecule or peptidomimetic, or an inhibitory nucleic acid that targets genomic or expressed nucleic acids (e.g., mRNA) encoding the target molecule, or a vector that encodes an inhibitory nucleic acid.
  • the compound can reduce the expression or bioavailability of the target molecule.
  • the inhibition can be competitive, non-competitive, uncompetitive, or product inhibition.
  • an inhibitor can directly inhibit the target molecule, an inhibitor can inhibit another factor in a pathway that leads to induction, persistence, or amplification of the target molecule’s expression, or a combination thereof.
  • the therapeutic agents can be and are also referred to herein as inhibitors.
  • the therapeutic agent is a protein binder that specifically binds to the target molecule, or a ligand or receptor thereof important for activity of the target molecule.
  • the protein binder is an antibody.
  • Antibodies include not only intact antibodies, but also antibody fragments and antigen-binding components thereof, and fusion proteins including antigen binding fragments that are capable of immuno-specifically binding to the target molecule (or its counterpart ligand or receipt) .
  • the antibodies can be a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a recombinant antibody, an antigen-binding antibody fragment, a single chain antibody, a monomeric antibody, a diabody, a triabody, a tetrabody, a Fab fragment, an IgD antibody, an IgE antibody, an IgM antibody, an IgG1 antibody, an IgG2 antibody, an IgG3 antibody, and an IgG4 antibody, or a fragment thereof, and fusion proteins formed therefrom.
  • the antibodies and antigen binding fragments can be monospecific, bispecific, trispecific or multispecific.
  • the inhibitor can be a functional nucleic acid.
  • Functional nucleic acids are nucleic acid molecules that have a specific function, such as binding a target molecule or catalyzing a specific reaction. As discussed in more detail below, functional nucleic acid molecules can be divided into the following non-limiting categories: antisense molecules, siRNA, miRNA, aptamers, ribozymes, triplex forming molecules, RNAi, and external guide sequences.
  • the functional nucleic acid molecules can act as effectors, inhibitors, modulators, and stimulators of a specific activity possessed by a target molecule, or the functional nucleic acid molecules can possess a de novo activity independent of any other molecules.
  • Functional nucleic acid molecules can interact with any macromolecule, such as DNA, RNA, polypeptides, or carbohydrate chains.
  • functional nucleic acids can interact with the mRNA or the genomic DNA of a target polypeptide or they can interact with the polypeptide itself.
  • functional nucleic acids are designed to interact with other nucleic acids based on sequence homology between the target molecule and the functional nucleic acid molecule.
  • the specific recognition between the functional nucleic acid molecule and the target molecule is not based on sequence homology between the functional nucleic acid molecule and the target molecule, but rather is based on the formation of tertiary structure that allows specific recognition to take place.
  • the compositions can include one or more functional nucleic acids designed to reduce expression of the target molecule’s gene, or a gene product thereof.
  • the functional nucleic acid or polypeptide can be designed to target and reduce or inhibit expression or translation of target molecule’s mRNA; or to reduce or inhibit expression, reduce activity, or increase degradation of target molecule protein.
  • the composition includes a vector suitable for in vivo expression of the functional nucleic acid.
  • Examples of functional nucleic acids include, but are not limited to, antisense oligonucleotides, siRNA, shRNA, miRNA, external guide sequences. External guide sequences (EGSs) , ribozymes, aptamers, and CRISPR/Cas technology.
  • EGSs External guide sequences
  • ribozymes ribozymes
  • aptamers aptamers
  • CRISPR/Cas technology CRISPR/Cas technology
  • compositions can be administered systemically.
  • Drugs can be formulated for immediate release, extended release, or modified release.
  • a delayed release dosage form is one that releases a drug (or drugs) at a time other than promptly after administration.
  • An extended release dosage form is one that allows at least a twofold reduction in dosing frequency as compared to that drug presented as a conventional dosage form (e.g. as a solution or prompt drug-releasing, conventional solid dosage form) .
  • a modified release dosage form is one for which the drug release characteristics of time course and/or location are chosen to accomplish therapeutic or convenience objectives not offered by conventional dosage forms such as solutions, ointments, or promptly dissolving dosage forms. Delayed release and extended release dosage forms and their combinations are types of modified release dosage forms.
  • Formulations are typically prepared using a pharmaceutically acceptable “carrier” composed of materials that are considered safe and effective and may be administered to an individual without causing undesirable biological side effects or unwanted interactions.
  • the “carrier” is all components present in the pharmaceutical formulation other than the active ingredient or ingredients.
  • carrier includes, but is not limited to, diluents, binders, lubricants, disintegrators, fillers, and coating compositions.
  • Carrier also includes all components of the coating composition which may include plasticizers, pigments, colorants, stabilizing agents, and glidants.
  • the delayed release dosage formulations may be prepared as described in references such as “Pharmaceutical dosage form tablets” , eds. Liberman et al.
  • the compound can be administered to a subject with or without the aid of a delivery vehicle.
  • Appropriate delivery vehicles for the compounds are known in the art and can be selected to suit the particular active agent.
  • the active agent (s) is incorporated into or encapsulated by, or bound to, a nanoparticle, microparticle, micelle, synthetic lipoprotein particle, or carbon nanotube.
  • the compositions can be incorporated into a vehicle such as polymeric particles which provide controlled release of the active agent (s) .
  • release of the drug (s) is controlled by diffusion of the active agent (s) out of the particles and/or degradation of the polymeric particles by hydrolysis and/or enzymatic degradation.
  • Suitable polymers include ethylcellulose and other natural or synthetic cellulose derivatives. Polymers which are slowly soluble and form a gel in an aqueous environment, such as hydroxypropyl methylcellulose or polyethylene oxide, may also be suitable as materials for drug containing particles or particles.
  • polymers include, but are not limited to, polyanhydrides, poly (ester anhydrides) , polyhydroxy acids, such as polylactide (PLA) , polyglycolide (PGA) , poly (lactide-co-glycolide) (PLGA) , poly-3-hydroxybut rate (PHB) and copolymers thereof, poly-4-hydroxybutyrate (P4HB) and copolymers thereof, polycaprolactone and copolymers thereof, and combinations thereof.
  • both agents are incorporated into the same particles and are formulated for release at different times and/or over different time periods. For example, in some forms, one of the agents is released entirely from the particles before release of the second agent begins. In other forms, release of the first agent begins followed by release of the second agent before the all of the first agent is released. In still other forms, both agents are released at the same time over the same period of time or over different periods of time.
  • Suitable pharmaceutically acceptable carriers and excipients are generally recognized as safe (GRAS) , and may be administered to an individual without causing undesirable biological side effects or unwanted interactions.
  • Representative carriers and excipients include solvents (including buffers) , diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agents, viscosity modifiers, tonicity agents, and stabilizing agents, and a combination thereof.
  • Excipients can be added to a liquid or solid pharmaceutical composition (for in vivo or in vitro applications) to assist in sterility, stability (e.g. shelf-life) , integration, and to adjust and/or maintain pH or isotonicity of the inhibitors in the pharmaceutical composition, such as diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agents, viscosity modifiers, tonicity agents, and stabilizing agents, and a combination thereof.
  • diluents pH modifying agents, preservatives, antioxidants, suspending agents, wetting agents, viscosity modifiers, tonicity agents, and stabilizing agents, and a combination thereof.
  • Suitable oral dosage forms include tablets, capsules, solutions, suspensions, syrups, and lozenges. Tablets can be made using compression or molding techniques well known in the art. Gelatin or non-gelatin capsules can prepared as hard or soft capsule shells, which can encapsulate liquid, solid, and semi-solid fill materials, using techniques well known in the art.
  • suitable coating materials include, but are not limited to, cellulose polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate and hydroxypropyl methylcellulose acetate succinate; polyvinyl acetate phthalate, acrylic acid polymers and copolymers, and methacrylic resins that are commercially available under the trade name (Roth Pharma, Westerstadt, Germany) , Zein, shellac, and polysaccharides.
  • cellulose polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate and hydroxypropyl methylcellulose acetate succinate
  • polyvinyl acetate phthalate acrylic acid polymers and copolymers
  • methacrylic resins that are commercially available under the trade name (Roth Pharma, Westerstadt, Germany) , Zein, shellac, and poly
  • the coating material may contain conventional carriers such as plasticizers, pigments, colorants, glidants, stabilization agents, pore formers and surfactants.
  • Optional pharmaceutically acceptable excipients present in the drug-containing tablets, beads, granules or particles include, but are not limited to, diluents, binders, lubricants, disintegrants, colorants, stabilizers, and surfactants.
  • Diluents also termed “fillers, " are typically necessary to increase the bulk of a solid dosage form so that a practical size is provided for compression of tablets or formation of beads and granules.
  • Suitable diluents include, but are not limited to, , dicalcium phosphate dihydrate, calcium sulfate, lactose, sucrose, mannitol, sorbitol, cellulose, microcrystalline cellulose, kaolin, sodium chloride, dry starch, hydrolyzed starches, pregelatinized starch, silicone dioxide, titanium oxide, magnesium aluminum silicate and powder sugar.
  • Binders are used to impart cohesive qualities to a solid dosage formulation and thus ensure that a tablet or bead or granule remains intact after the formation of the dosage forms.
  • Suitable binder materials include, but are not limited to, starch, pregelatinized starch, gelatin, sugars (including sucrose, glucose, dextrose, lactose and sorbitol) , polyethylene glycol, waxes, natural and synthetic gums such as acacia, tragacanth, sodium alginate, cellulose, including hydorxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, and veegum, and synthetic polymers such as acrylic acid and methacrylic acid copolymers, methacrylic acid copolymers, methyl methacrylate copolymers, aminoalkyl methacrylate copolymers, polyacrylic acid/polymethacrylic acid and polyvinylpyrrolidone.
  • Lubricants are used to facilitate tablet manufacture.
  • suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, glycerol behenate, polyethylene glycol, talc, and mineral oil.
  • Disintegrants are used to facilitate dosage form disintegration or "breakup" after administration, and generally include, but are not limited to, starch, sodium starch glycolate, sodium carboxymethyl starch, sodium carboxymethylcellulose, hydroxypropyl cellulose, pregelatinized starch, clays, cellulose, alginine, gums or cross-linked polymers, such as cross-linked PVP (Polyplasdone XL from GAF Chemical Corp) .
  • PVP Polyplasdone XL from GAF Chemical Corp
  • Stabilizers are used to inhibit or retard drug decomposition reactions which include, by way of example, oxidative reactions.
  • Surfactants may be anionic, cationic, amphoteric or nonionic surface-active agents.
  • Suitable anionic surfactants include, but are not limited to, those containing carboxylate, sulfonate and sulfate ions.
  • anionic surfactants include sodium, potassium, ammonium of long chain alkyl sulfonates and alkyl aryl sulfonates such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium bis- (2-ethylthioxyl) -sulfosuccinate; and alkyl sulfates such as sodium lauryl sulfate.
  • Cationic surfactants include, but are not limited to, quaternary ammonium compounds such as benzalkonium chloride, benzethonium chloride, cetrimonium bromide, stearyl dimethylbenzyl ammonium chloride, polyoxyethylene and coconut amine.
  • nonionic surfactants include ethylene glycol monostearate, propylene glycol myristate, glyceryl monostearate, glyceryl stearate, polyglyceryl-4-oleate, sorbitan acylate, sucrose acylate, PEG-150 laurate, PEG-400 monolaurate, polyoxyethylene monolaurate, polysorbates, polyoxyethylene octylphenylether, PEG-1000 cetyl ether, polyoxyethylene tridecyl ether, polypropylene glycol butyl ether, 401, stearoyl monoisopropanolamide, and polyoxyethylene hydrogenated tallow amide.
  • amphoteric surfactants include sodium N-dodecyl-. beta. -alanine, sodium N-lauryl-. beta. -iminodipropionate, myristoamphoacetate, lauryl betaine and lauryl sulfobetaine.
  • the tablets, beads granules or particles may also contain minor amount of nontoxic auxiliary substances such as wetting or emulsifying agents, dyes, pH buffering agents, and preservatives.
  • the extended release formulations are generally prepared as diffusion or osmotic systems, for example, as described in “Remington –The science and practice of pharmacy” (20th ed., Lippincott Williams &Wilkins, Baltimore, MD, 2000) .
  • a diffusion system typically consists of two types of devices, reservoir and matrix, and is well known and described in the art.
  • the matrix devices are generally prepared by compressing the drug with a slowly dissolving polymer carrier into a tablet form.
  • the three major types of materials used in the preparation of matrix devices are insoluble plastics, hydrophilic polymers, and fatty compounds.
  • Plastic matrices include, but not limited to, methyl acrylate-methyl methacrylate, polyvinyl chloride, and polyethylene.
  • Hydrophilic polymers include, but are not limited to, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and carbopol 934, polyethylene oxides.
  • Fatty compounds include, but are not limited to, various waxes such as carnauba wax and glyceryl tristearate.
  • extended release formulations can be prepared using osmotic systems or by applying a semi-permeable coating to the dosage form.
  • the desired drug release profile can be achieved by combining low permeable and high permeable coating materials in suitable proportion.
  • the devices with different drug release mechanisms described above could be combined in a final dosage form having single or multiple units.
  • Examples of multiple units include multilayer tablets, capsules containing tablets, beads, granules, etc.
  • An immediate release portion can be added to the extended release system by means of either applying an immediate release layer on top of the extended release core using coating or compression process or in a multiple unit system such as a capsule containing extended and immediate release beads.
  • Extended release tablets containing hydrophilic polymers are prepared by techniques commonly known in the art such as direct compression, wet granulation, or dry granulation processes. Their formulations usually incorporate polymers, diluents, binders, and lubricants as well as the active pharmaceutical ingredient.
  • the usual diluents include inert powdered substances such as any of many different kinds of starch, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
  • Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives are also useful.
  • Typical tablet binders include substances such as starch, gelatin and sugars such as lactose, fructose, and glucose. Natural and synthetic gums, including acacia, alginates, methylcellulose, and polyvinylpyrrolidine can also be used. Polyethylene glycol, hydrophilic polymers, ethylcellulose and waxes can also serve as binders.
  • a lubricant is necessary in a tablet formulation to prevent the tablet and punches from sticking in the die. The lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.
  • Extended release tablets containing wax materials are generally prepared using methods known in the art such as a direct blend method, a congealing method, and an aqueous dispersion method.
  • a congealing method the drug is mixed with a wax material and either spray-congealed or congealed and screened and processed.
  • Delayed release formulations are created by coating a solid dosage form with a film of a polymer which is insoluble in the acid environment of the stomach, and soluble in the neutral environment of small intestines.
  • the delayed release dosage units can be prepared, for example, by coating a drug or a drug-containing composition with a selected coating material.
  • the drug-containing composition may be, e.g., a tablet for incorporation into a capsule, a tablet for use as an inner core in a "coated core” dosage form, or a plurality of drug-containing beads, particles or granules, for incorporation into either a tablet or capsule.
  • Preferred coating materials include bioerodible, gradually hydrolyzable, gradually water-soluble, and/or enzymatically degradable polymers, and may be conventional "enteric" polymers.
  • Enteric polymers become soluble in the higher pH environment of the lower gastrointestinal tract or slowly erode as the dosage form passes through the gastrointestinal tract, while enzymatically degradable polymers are degraded by bacterial enzymes present in the lower gastrointestinal tract, particularly in the colon.
  • Suitable coating materials for effecting delayed release include, but are not limited to, cellulosic polymers such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxymethyl cellulose, hydroxypropyl methyl cellulose, hydroxypropyl methyl cellulose acetate succinate, hydroxypropylmethyl cellulose phthalate, methylcellulose, ethyl cellulose, cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate and carboxymethylcellulose sodium; acrylic acid polymers and copolymers, preferably formed from acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate, and other methacrylic resins that are commercially available under the tradename (Rohm Pharma; Westerstadt, Germany) , including L30D-55 and L100-55 (soluble at pH 5.5 and above) , L-100 (soluble at pH 6.0 and above) , S
  • the preferred coating weights for particular coating materials may be readily determined by those skilled in the art by evaluating individual release profiles for tablets, beads and granules prepared with different quantities of various coating materials. It is the combination of materials, method and form of application that produce the desired release characteristics, which one can determine only from the clinical studies.
  • the coating composition may include conventional additives, such as plasticizers, pigments, colorants, stabilizing agents, glidants, etc.
  • a plasticizer is normally present to reduce the fragility of the coating, and will generally represent about 10 wt. %to 50 wt. %relative to the dry weight of the polymer.
  • typical plasticizers include polyethylene glycol, propylene glycol, triacetin, dimethyl phthalate, diethyl phthalate, dibutyl phthalate, dibutyl sebacate, triethyl citrate, tributyl citrate, triethyl acetyl citrate, castor oil and acetylated monoglycerides.
  • a stabilizing agent is preferably used to stabilize particles in the dispersion.
  • Typical stabilizing agents are nonionic emulsifiers such as sorbitan esters, polysorbates and polyvinylpyrrolidone. Glidants are recommended to reduce sticking effects during film formation and drying, and will generally represent approximately 25 wt. %to 100 wt. %of the polymer weight in the coating solution.
  • One effective glidant is talc.
  • Other glidants such as magnesium stearate and glycerol monostearates may also be used.
  • Pigments such as titanium dioxide may also be used.
  • Small quantities of an anti-foaming agent such as a silicone (e.g., simethicone) , may also be added to the coating composition.
  • Such methods include, but are not limited to, the following: coating a drug or drug-containing composition with an appropriate coating material, typically although not necessarily incorporating a polymeric material, increasing drug particle size, placing the drug within a matrix, and forming complexes of the drug with a suitable complexing agent.
  • a method for preparing extended release tablets is by compressing a drug-containing blend, e.g., blend of granules, prepared using a direct blend, wet-granulation, or dry-granulation process.
  • Extended release tablets may also be molded rather than compressed, starting with a moist material containing a suitable water-soluble lubricant. However, tablets are preferably manufactured using compression rather than molding.
  • a preferred method for forming extended release drug-containing blend is to mix drug particles directly with one or more excipients such as diluents (or fillers) , binders, disintegrants, lubricants, glidants, and colorants.
  • a drug-containing blend may be prepared by using wet-granulation or dry-granulation processes.
  • Beads containing the active agent may also be prepared by any one of a number of conventional techniques, typically starting from a fluid dispersion.
  • a typical method for preparing drug-containing beads involves dispersing or dissolving the active agent in a coating suspension or solution containing pharmaceutical excipients such as polyvinylpyrrolidone, methylcellulose, talc, metallic stearates, silicone dioxide, plasticizers or the like.
  • the admixture is used to coat a bead core such as a sugar sphere (or so-called "non-pareil” ) having a size of approximately 60 to 20 mesh.
  • An alternative procedure for preparing drug beads is by blending drug with one or more pharmaceutically acceptable excipients, such as microcrystalline cellulose, lactose, cellulose, polyvinyl pyrrolidone, talc, magnesium stearate, a disintegrant, etc., extruding the blend, spheronizing the extrudate, drying and optionally coating to form the immediate release beads.
  • excipients such as microcrystalline cellulose, lactose, cellulose, polyvinyl pyrrolidone, talc, magnesium stearate, a disintegrant, etc.
  • the disclosed composition is formulated for direct application on the mucous membrane generally contain a dermatologically acceptable carrier that is suitable for application to the mucous membrane, has good aesthetic properties, is compatible with the active agents and any other components, and will not cause any untoward safety or toxicity concerns.
  • the carrier can be in a wide variety of forms.
  • emulsion carriers including, but not limited to, oil-in-water, water-in-oil, water-in-oil-in-water, and oil-in-water-in-silicone emulsions, are useful herein. These emulsions can cover a broad range of viscosities, e.g., from about 100 cps to about 200,000 cps. These emulsions can also be delivered in the form of sprays using either mechanical pump containers or pressurized aerosol containers using conventional propellants. These carriers can also be delivered in the form of a mousse or a transdermal patch.
  • suitable topical carriers include anhydrous liquid solvents such as oils, alcohols, and silicones (e.g., mineral oil, ethanol isopropanol, dimethicone, cyclomethicone, and the like) ; aqueous-based single phase liquid solvents (e.g., hydro-alcoholic solvent systems, such as a mixture of ethanol and/or isopropanol and water) ; and thickened versions of these anhydrous and aqueous-based single phase solvents (e.g. where the viscosity of the solvent has been increased to form a solid or semi-solid by the addition of appropriate gums, resins, waxes, polymers, salts, and the like) .
  • anhydrous liquid solvents such as oils, alcohols, and silicones (e.g., mineral oil, ethanol isopropanol, dimethicone, cyclomethicone, and the like)
  • aqueous-based single phase liquid solvents e.g., hydro-alcoholic solvent systems,
  • topical carrier systems useful in the present formulations are described in the following four references all of which are incorporated herein by reference in their entirety: “Sun Products Formulary” Cosmetics &Toiletries, vol. 105, pp. 122-139 (December 1990) ; “Sun Products Formulary, ” Cosmetics &Toiletries, vol. 102, pp. 117-136 (March 1987) ; U.S. Pat. No. 5,605,894 to Blank et al., and U.S. Pat. No. 5,681,852 to Bissett.
  • Formulations for direct application on the mucous membrane may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed, sustained, pulsed, controlled, targeted and programmed release formulations.
  • the disclosed composition may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active agents.
  • the disclosed composition can be formulated for Parenteral administration.
  • the disclosed composition can be formulated in a form suitable for administration directly into the blood stream, into muscle, or into an internal organ. Suitable routes for such parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, epidural, intracerebroventricular, intraurethral, intrasternal, intracranial, intramuscular, and subcutaneous delivery. Suitable means for parenteral administration include needle (including microneedle) injectors, needle free injectors, and infusion techniques.
  • the disclosed composition can be formulated in a form suitable for intramuscular administration, intravenous administration, intraperitoneal administration, or subcutaneous administration, or a combination thereof.
  • the disclosed composition described herein can be in aqueous solutions which can contain excipients such as salts, carbohydrates and buffering agents (e.g., from about pH 6.5 to about pH 8.0, from about pH 6.5 to about pH 7.4, from about pH 6.5 to about pH 7.0, from about pH 7.0 to pH 8.0, or from about pH 7.0 to about pH 7.4) , but, for some applications, they may be more suitably formulated as a sterile aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen free water.
  • excipients such as salts, carbohydrates and buffering agents (e.g., from about pH 6.5 to about pH 8.0, from about pH 6.5 to about pH 7.4)
  • buffering agents e.g., from about pH 6.5 to about pH 8.0, from about pH 6.5 to about pH 7.4, from about pH 6.5 to about pH 7.0, from about pH 7.0 to pH 8.0, or from about pH 7.0 to about pH 7.4
  • the disclosed composition may be a solution, a suspension, or an emulsion.
  • the disclosed composition can include one or more physiologically compatible buffers, such as a phosphate buffers.
  • physiologically compatible buffers such as a phosphate buffers.
  • a suitable saline content and pH for an aqueous carrier for administration e.g., from about pH 6.5 to about pH 8.0, from about pH 6.5 to about pH 7.4, from about pH 6.5 to about pH 7.0, from about pH 7.0 to pH 8.0, or from about pH 7.0 to about pH 7.4 .
  • the disclosed composition for parenteral administration may include one or more suspending agents, such as cellulose derivatives, sodium alginate, polyvinylpyrrolidone, gum tragacanth, or lecithin.
  • the liquid compositions may also include one or more preservatives, such as ethyl or n-propyl p-hydroxybenzoate.
  • the disclosed composition may contain one or more solvents that are low toxicity organic (i.e., nonaqueous) class 3 residual solvents, such as ethanol, acetone, ethyl acetate, tetrahydofuran, ethyl ether, and propanol, and a combination thereof.
  • Solvents such as freon, alcohol, glycol, polyglycol, or fatty acid, can also be included in the liquid composition.
  • the disclosed composition may also contain minor amounts of polymers, surfactants, or other pharmaceutically acceptable excipients known to those in the art.
  • the disclosed composition is formulated typically under sterile conditions, for example, by lyophilisation, which can be accomplished using standard pharmaceutical techniques known to those skilled in the art.
  • the disclosed composition may be formulated to provide immediate and/or modified release.
  • Modified release formulations include delayed, sustained, pulsed, controlled, targeted and programmed release formulations.
  • the subject to be treated can have a disease, disorder, or condition such as but not limited to, cancer, an immune system disorder such autoimmune disease, an inflammatory disease, a neuronal disorder, HIV/AIDS, diabetes, a cardiovascular disease, an infectious disease, or combinations thereof.
  • a disease, disorder, or condition such as but not limited to, cancer, an immune system disorder such autoimmune disease, an inflammatory disease, a neuronal disorder, HIV/AIDS, diabetes, a cardiovascular disease, an infectious disease, or combinations thereof.
  • the methods treat or prevent cancer. In some forms, the methods treat or prevent metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) . In some forms, the methods treat or prevent colorectal cancer (CRC) .
  • MASLD-HCC metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma
  • CRC colorectal cancer
  • the methods treat or prevent cancer or other proliferative disease or disorder in a subject identified as having, or at risk of having cancer or other proliferative disease or disorder.
  • Cancer is a disease of genetic instability, allowing a cancer cell to acquire the hallmarks proposed by Hanahan and Weinberg, including (i) self-sufficiency in growth signals; (ii) insensitivity to anti-growth signals; (iii) evading apoptosis; (iv) sustained angiogenesis; (v) tissue invasion and metastasis; (vi) limitless replicative potential; (vii) reprogramming of energy metabolism; and (viii) evading immune destruction (Cell., 144: 646–674, (2011) ) .
  • Tumors which can be treated in accordance with the disclosed methods, are classified according to the embryonic origin of the tissue from which the tumor is derived.
  • Carcinomas are tumors arising from endodermal or ectodermal tissues such as skin or the epithelial lining of internal organs and glands.
  • Sarcomas which arise less frequently, are derived from mesodermal connective tissues such as bone, fat, and cartilage.
  • the leukemias and lymphomas are malignant tumors of hematopoietic cells of the bone marrow. Leukemias proliferate as single cells, whereas lymphomas tend to grow as tumor masses. Malignant tumors may show up at numerous organs or tissues of the body to establish a cancer.
  • Table 5 The disclosed compositions and methods can be used in the treatment of one or more cancers.
  • compositions and methods of treatment thereof are generally suited for treatment of carcinomas, sarcomas, lymphomas and leukemias.
  • the described compositions and methods are useful for treating, or alleviating subjects having benign or malignant tumors by delaying or inhibiting the growth/proliferation or viability of tumor cells in a subject, reducing the number, growth or size of tumors, inhibiting or reducing metastasis of the tumor, and/or inhibiting or reducing symptoms associated with tumor development or growth.
  • the types of cancer that can be treated with the provided compositions and methods include, but are not limited to, cancers such as vascular cancer such as multiple myeloma, adenocarcinomas and sarcomas, of bone, bladder, brain, breast, cervical, colorectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, and uterine.
  • cancers such as vascular cancer such as multiple myeloma, adenocarcinomas and sarcomas, of bone, bladder, brain, breast, cervical, colorectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, and uterine.
  • vascular cancer such as multiple myeloma, adenocarcinomas and sarcomas
  • the target cancer is a solid tumor.
  • the compositions are used to treat multiple cancer types concurrently.
  • tumor cells include, but are not limited to, tumor cells of cancers, including leukemias including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome, chronic leukemias such as, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as, but not limited to, Hodgkin’s disease, non-Hodgkin’s disease; multiple myelomas such as, but not limited to, smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma and extramedul
  • compositions and methods for treating metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) or colorectal cancer (CRC) can be used in combination with other therapeutic agents or treatment modalities.
  • MASLD-HCC metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma
  • CRC colorectal cancer
  • compositions and methods for treating metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma can be used in combination with other therapeutic agents or treatment modalities such as chemotherapy, stem-cell transplantation, or immunotherapy.
  • the pharmaceutical compositions and other therapeutic agents are administered separately through the same route of administration. In other forms, the pharmaceutical compositions and other therapeutic agents are administered separately through different routes of administration.
  • the combinations can be administered either concomitantly (e.g., as an admixture) , separately but simultaneously (e.g., via separate intravenous lines into the same subject; one agent is given orally while the other agent is given by infusion or injection, etc., ) , or sequentially (e.g., one agent is given first followed by the second) .
  • compositions and methods described herein may be used as a first therapy, second therapy, third therapy, or combination therapy with other types of therapies known in the art, such as chemotherapy, surgery, radiation, gene therapy, immunotherapy, bone marrow transplantation, stem cell transplantation, targeted therapy, cryotherapy, ultrasound therapy, photodynamic therapy, radio-frequency ablation or the like, in an adjuvant setting or a neoadjuvant setting.
  • therapies known in the art, such as chemotherapy, surgery, radiation, gene therapy, immunotherapy, bone marrow transplantation, stem cell transplantation, targeted therapy, cryotherapy, ultrasound therapy, photodynamic therapy, radio-frequency ablation or the like, in an adjuvant setting or a neoadjuvant setting.
  • the disclosed pharmaceutical compositions and/or other therapeutic agents, procedures or modalities can be administered during periods of active disease, or during a period of remission or less active disease.
  • the pharmaceutical compositions can be administered before the additional treatment, concurrently with the treatment, post-treatment, or during remission of the disease or disorder.
  • the disclosed pharmaceutical compositions and the additional therapeutic agents e.g., second or third agent
  • the disclosed pharmaceutical compositions and the additional therapeutic agents can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the administered amount or dosage of the disclosed pharmaceutical composition, the additional therapeutic agent (e.g., second or third agent) , or all is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy (e.g., required to achieve the same therapeutic effect) .
  • examples of additional therapeutic agents include other conventional therapies known in the art for treating the desired disease, disorder or condition.
  • the therapeutic agent is one or more other targeted therapies (e.g., a targeted cancer therapy) and/or immune-checkpoint blockage agents (e.g., anti-LAG-3, anti CTLA 4, anti PD1, and/or anti PDL1 agents such as antibodies) .
  • a treatment regimen can include one or multiple administrations of the compositions including an effective amount of one or more of the compounds for achieving a desired physiological change, including administering to an animal, such as a mammal, especially a human being, an effective amount of the compositions to treat the disease or symptom thereof, or to produce the physiological change.
  • the effective amount or therapeutically effective amount of a pharmaceutical compositions can be a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of a disease or disorder or to otherwise provide a desired pharmacologic and/or physiologic effect, for example, reducing, inhibiting, or reversing one or more of the underlying pathophysiological mechanisms underlying a disease or disorder, such as PCAD.
  • the desired physiological change could include improvement in one or more symptoms of a disease or condition treated herein, such as improvement in breathing and exercise capacity or improved sensitivity to irritants or drop in albuterol use or improved vocal cord function or reduction in cough in the subject.
  • the amount administered when administrating the pharmaceutical composition, can be expressed as the amount effective to achieve a desired effect in the recipient.
  • the effective amount of the pharmaceutical composition will vary based on the active agent and from subject to subject, depending on the species, age, weight and general condition of the subject, the severity of the disorder being treated, and its mode of administration. Thus, it is not possible to specify an exact amount for every pharmaceutical composition. However, an appropriate amount can be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. For example, effective dosages and schedules for administering the pharmaceutical composition can be determined empirically. In some forms, the dosage ranges for the administration of the composition are those large enough to resolve mucosal hyperreactivity throughout the respiratory tract.
  • the dosage is not so large as to cause adverse side effects, such as unwanted cross-reactions, anaphylactic reactions, and the like.
  • the dosage will vary with the age, condition, and sex of the patient, route of administration, whether other drugs are included in the regimen, and the type, stage, and location of the disease to be treated.
  • the dosage can be adjusted by the individual physician in the event of any counter-indications.
  • the effective dosage of the composition can increase or decrease over the course of a particular treatment. Changes in dosage can result and become apparent from the results of diagnostic assays.
  • Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. Guidance can be found in the literature for appropriate dosages for given classes of pharmaceutical products. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the subject or patient. Persons of ordinary skill can determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages can vary depending on the relative potency of individual pharmaceutical compositions, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models.
  • examples of daily dosages of the compounds described herein which can be used are an effective amount within the dosage range of about 0.001 mg to about 2 mg per kilogram of body weight, about 0.001 mg to about 5 mg per kilogram of body weight, about 0.001 mg to about 10 mg per kilogram of body weight, about 0.001 mg to about 20 mg per kilogram of body weight, about 0.001 mg to about 50 mg per kilogram of body weight, about 0.001 mg to about 100 mg per kilogram of body weight, about 0.001 mg to about 200 mg per kilogram of body weight, or about 0.001 mg to about 300 mg per kilogram of body weight.
  • examples of daily dosages are an effective amount within the dosage range of about 0.1 mg to about 10 mg, or about 0.1 mg to about 20 mg, or about 0.1 mg to about 30 mg, or about 0.1 mg to about 40 mg, or about 0.1 mg to about 50 mg, or about 0.1 mg to about 60 mg, or about 0.1 mg to about 70 mg, or about 0.1 mg to about 80 mg, or about 0.1 mg to about 90 mg, or about 0.1 mg to about 100 mg, or about 0.1 mg to about 200 mg, or about 0.1 mg to about 300 mg, or about 0.1 mg to about 400 mg, or about 0.1 mg to about 500 mg, or about 0.1 mg to about 600 mg, or about 0.1 mg to about 700 mg, or about 0.1 mg to about 800 mg, or about 0.1 mg to about 900 mg, or about 0.1 mg to about 1 g, or about 20 mg to 300 mg, or about 20 mg to 500 mg, or about 20 mg to 700 mg, or about 20 mg to 1000 mg, or about 50 mg to 1500 mg, or about 50 mg to
  • Exemplary fixed daily doses include about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 12 mg, about 15 mg, about 18 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg, about 1200 mg, about 1500 mg, or about 2000 mg, independently of body weight.
  • pediatric patients may require smaller dosages, and depending on the severity of the disease and condition of the patient, dosages may vary.
  • the concentration of the compounds described herein may be about 0.01 mg/ml to about 0.1 mg/ml or about 0.1 mg/ml to about 1 mg/ml, but can also be about 1 mg/ml to about 10 mg/ml or about 10 mg/ml to about 100 mg/ml.
  • the liquid formulation could be a solution or a suspension.
  • the concentration when formulated as a solid, for example as a tablet or as a powder for inhalation, the concentration, expressed as the weight of a compound divided by total weight, will typically be about 0.01%to about 0.1%, about 0.1%to about 1%, about 1%to about 10%, about 10%to about 20%, about 20%to about 40%, about 40%to about 60%, about 60%to about 80%, or about 80%to about 100%.
  • administration of the composition will be given as a long-term treatment regimen whereby pharmacokinetic steady state conditions will be reached.
  • the unit dosage is in a unit dosage form for intravenous injection. In some forms, the unit dosage is in a unit dosage form for oral administration. In some forms, the unit dosage is in a unit dosage form for inhalation. In some forms, the unit dosage is in a unit dosage form for subcutaneous injection.
  • Treatment can be continued for an amount of time sufficient to achieve one or more desired therapeutic goals.
  • the timing of the administration of the composition will also depend on the formulation and/or route of administration used.
  • the compound may be administered once daily, but may also be administered two, three or four times daily, or every other day, or once or twice per week.
  • the subject can be administered one or more treatments 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, days, weeks, or months apart.
  • Treatment can be continued for a desired period of time, and the progression of treatment can be.
  • administration is carried out every day of treatment, or every week, or every fraction of a week.
  • treatment regimens are carried out over the course of up to two, three, four or five days, weeks, or months, or for up to 6 months, or for more than 6 months, for example, up to one year, two years, three years, or up to five years.
  • the efficacy of administration of a particular dose of the pharmaceutical compositions can be determined by evaluating the aspects of the medical history, signs, symptoms, and objective laboratory tests that are known to be useful in evaluating the status of a subject in need for the treatment of a disease or condition discussed herein. These signs, symptoms, and objective laboratory tests will vary, depending upon the particular disease or condition being treated or prevented, as will be known to any clinician who treats such patients or a researcher conducting experimentation in this field.
  • Example 1 Probiotics mixture, prohep, is an adjuvant for low-dose sorafenib in metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma treatment through modulating gut microbiota.
  • Prohep is a proprietary probiotic mixture developed by the inventors of this application (IP01473) .
  • the modified formula was composed of Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium lactis and Streptococcus thermophilus in lyophilized powder, produced under GMP (Fukopharma, Finland) as previously reported [Zhang, F., et al., Probiotic Mixture Ameliorates a Diet-Induced MASLD/MASH Murine Model through the Regulation of Hepatic Lipid Metabolism and the Gut Microbiome. J Agric Food Chem, 2024.72 (15) : p. 8536-8549] .
  • mice Two weeks old male C57BL/6J mice (Center of Comparative Medicine Research, The University of Hong Kong) were maintained under a controlled environment (23 ⁇ 1 °C, 50–60%humidity, 12 h light/dark cycles) with foster mothers. All animal experimental procedures were approved by the Committee on the Use of Live Animals in Teaching and Research at the University of Hong Kong (CULATR 5544-20) and received humane care according to the criteria outlined in the “Guide for the Care and Use of Laboratory Animals. ” The mice were intraperitoneally injected with a single dose of diethylnitrosamine (DEN; 25 mg/kg) .
  • DEN diethylnitrosamine
  • mice were provided with high-fat high-cholesterol (HFD) diet (containing 60%fat, 20%carbohydrates, 19.5%proteins, and 0.5%cholesterol, TP26304, trophic diet, Nantong, China) until sacrifice.
  • HFD high-fat high-cholesterol
  • TP26304 trophic diet, Nantong, China
  • the mice were then randomly distributed and trained for one week for voluntary oral MediGel sucralose gel administration.
  • the control group (Con) and Prohep group (Pro) were given daily either 0.25 g of MediGel sucralose (ClearH2O, ME, US) , which served as a control gel, or 7 ⁇ 10 9 CFU per mice of Prohep in a control gel from until sacrifice.
  • Sorafenib group (Sora) was the positive control and was supplemented (30mg/kg) starting from week 24.
  • daily Sorafenib (30mg/kg) was given in combination with daily Prohep (7 ⁇ 10 9 CFU per mouse) from the age of 24 weeks until sacrifice.
  • mice received daily Prohep for four weeks from the age of 20 weeks before the combined usage of Prohep and Sorafenib until sacrifice. The supplementation of Sorafenib and Prohep were given in the morning (10 AM) and afternoon (3 PM) respectively.
  • Fresh mouse liver tissues were processed and embedded in paraffin or directly embedded with an optimal cutting temperature compound to make frozen sections (Sakura Finetek USA, lnc., Torrance) .
  • the paraffin sections were prepared in 5 ⁇ m thickness, subsequently deparaffinized, and rehydrated in xylene and graded ethanol.
  • the sections were stained with a hematoxylin and eosin (H&E) kit (BASO, Wuhan, China) according to the manufacturer's instructions to evaluate the morphology and hepatic pathology.
  • H&E hematoxylin and eosin
  • the MASLD activity score was used to evaluate steatosis, lobular inflammation, and hepatocyte ballooning.
  • steatosis 0: no foci; 1: ⁇ 2 foci per 200 ⁇ field; 2: 2–4 foci per 200 ⁇ field; 3: >4 foci per 200 ⁇ field.
  • hepatocyte ballooning 0: none; 1: few balloon cells; 1: many balloon cells.
  • Sirius red staining (Abcam, Cambridge, UK) was performed to evaluate the degree of liver collagen deposition and fibrosis, and the degree of liver fibrosis was quantified by calculating the percentage of the positive red staining area over the total colon area measured.
  • Oil red O (ORO) staining (BASO, Wuhan, China) was used to evaluate the intrahepatic lipids. All sections were visualized with a biological microscope (Olympus, BX41) , and images were captured with a color digital camera (Olympus, DP72) .
  • a volume of 20 ⁇ L serum was diluted with deionized H 2 O for biochemical measurements.
  • Total cholesterol, low-density lipoprotein cholesterol (LDL-C) , high-density lipoprotein cholesterol (HDL-C) , alanine aminotransferase (ALT) , and aspartate aminotransferase (AST) were measured using a cobas c111 analyzer (Roche Diagnostics; Indianapolis, IN, USA) according to the manufacturer’s instructions.
  • Serum leptin was measured using Mouse/Rat Leptin Quantikine ELISA Kit (R&D Systems, Inc., USA) according to the manufacturer’s instructions.
  • TC hepatic total cholesterol
  • TG triglycerides
  • glycogen glycogen assay kit
  • liver tissues were homogenized with a RIPA buffer, and the levels of tumor necrosis factor- ⁇ (TNF- ⁇ ) , interleukin-17 (IL-17) , interleukin-6 (IL-6) , and interleukin-10 (IL-10) were determined via measuring the absorbance at 450 nm according to the manufacturer’s instructions (BioLegend, lnc., San Diego, CA) .
  • TNF- ⁇ tumor necrosis factor- ⁇
  • IL-17 interleukin-17
  • IL-6 interleukin-6
  • IL-10 interleukin-10
  • GSH Hepatic glutathione
  • SOD superoxide dismutase
  • RT-qPCR a reaction mixture containing an AceQ qPCR SYBR Green Master mix (Vazyme Biotech, China) , cDNA and primers was prepared. The sequences of primers used in this study are listed in Table 2) . Relative gene expression was normalized to GAPDH and calculated by the 2– ⁇ Ct method.
  • Protein extraction from liver tissues was performed using RIPA buffer supplemented with protease and phosphatase inhibitor from Sigma-Aldrich (St. Louis, MO, USA) .
  • the extracted protein was centrifuged at 13,000 ⁇ g for 15 minutes at 4°C.
  • the total protein content was quantified using the DC protein assay from Bio-Rad (Hercules, CA, USA) .
  • An equal amount of extracted protein was loaded onto 10%SDS-PAGE and transferred onto a polyvinylidene fluoride (PVDF) membrane.
  • PVDF polyvinylidene fluoride
  • the membrane was blocked with 5%non-fat milk or BSA and incubated overnight at 4°C with primary antibodies, including anti-FAS (1:1000) (ab271016) , PI3K signaling pathway panel (ab283852) , anti-FOXO3A (ab12162) , anti-Heme Oxygenase 1 antibody (HO-1) (ab13248) and anti-GAPDH mouse (ab9485) antibody from Abcam (Cambridge, UK) .
  • primary antibodies including anti-FAS (1:1000) (ab271016) , PI3K signaling pathway panel (ab283852) , anti-FOXO3A (ab12162) , anti-Heme Oxygenase 1 antibody (HO-1) (ab13248) and anti-GAPDH mouse (ab9485) antibody from Abcam (Cambridge, UK) .
  • HRP horseradish peroxidase
  • HRP conjugated with horseradish peroxidase horseradish peroxidase
  • proteins were visualized using enhanced chemiluminescence reagents from Bio-Rad (CA, USA) and imaged using the ChemiDox XRS+ imaging system from Bio-Rad (CA, USA) .
  • the intensity of the bands was quantified using ImageJ software from NIH (Bethesda, MD, USA) , and normalization was performed with reference to the intensity of GAPDH [Zhang et al. (2024) ] .
  • Short-chain fatty acids including acetate, propionate, butyrate, and valerate levels, were quantified in fecal samples using a modified protocol. Briefly, approximately 50 mg of fecal samples were homogenized in a 0.005 M sodium hydroxide buffer containing an internal standard (10 ⁇ g/mL acetic acid-d 4 , Cambridge Isotope Laboratories, USA) and centrifuged at 13, 200 x g for 20 minutes at 4°C. The liquid remaining after centrifugation was combined with a solution containing 1-propanol and pyridine in a 3: 2 ratio (volume/volume) , along with propyl chloroformate.
  • SCFAs Short-chain fatty acids
  • the electron energy used was -70 eV, and mass spectral data were collected in a full-scan mode within the m/z range of 30 to 600.
  • the levels of SCFAs were determined by creating calibration curves based on the peak areas of acetate, propionate, butyrate, and valerate against acetic acid-d 4 .
  • DNA of the fecal samples was extracted using a QIAamp PowerFecal Pro DNA kit (Qiagen, Germany) according to the manufacturer’s instructions and stored at -80 °C. The integrity of the DNA was confirmed through gel electrophoresis, and the purity and concentrations were checked using Thermo Scientific NanoDrop 2000/2000c spectrophotometers (Thermo Scientific, DE, USA) . After passing the quality check, the extracted samples were sent to Beijing Genomics Institute Hong Kong Co., Limited (BGI, Hong Kong) for sequencing using the DNBseq platform (BGI, Tianjin) .
  • the raw pair-end reads generated from metagenomic shotgun sequencing were aligned to the mouse reference genome GRCm39 using BWA-MEM for host contamination removal.
  • Adaptor regions, low-quality reads, and PCR duplicates were further filtered with an in-house script as previously described [Aron-Wisnewsky, J., et al., Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol, 2020.17 (5) : p. 279-297] .
  • R package phyloseq was used to aggregate the raw abundance table into a species-level counts per million (CPM) table.
  • CPM species-level counts per million
  • R package vegan was then used to determine alpha and beta diversity.
  • Kruskal-Wallis test following with Dunn’s test and PERMANOVA were applied for alpha diversity and beta diversity, respectively, to assess the overall composition difference between groups.
  • ANCOM-BC was used to compare the abundance of taxa between groups.
  • LDA Linear dis-criminant analysis
  • HUMAnN3 v3.7
  • KEGG Kyoto Encyclopaedia of Genes and Genomes
  • KEGG module were obtained by utilizing the HUMAnN3 regroup table function.
  • ANCOM-BC was used to assess the differential abundances (DA) of KO and the abundance of metabolic pathways. KO and pathway that met FDR ⁇ 0.05 were considered as differentially abundant.
  • Pro, Pro-Sora and PrePro-Sora groups showed significantly retarded tumor development as evidenced by significant decreases in the liver/body weight ratio, surface tumor counts, and surface maximum tumor sizes compared to Control group. While Sora group is effective in reducing liver/body weight ratio and surface maximum tumor sizes but not surface tumor counts. Prohep showed potential adjuvant effect as it greatly improved the therapeutic effect of Sorafenib. Of note, the tumor counts were significantly lower in the ProSora and PrePro-Sora groups compared to Sorafenib alone. The tumor burden was further confirmed by analysis of H&E staining of mouse liver sections (Fig. 1B) .
  • the mean maximum tumor size in Con, Pro, Sora, Pro-Sora and PrePro-Sora groups are 0.78, 0.23, 0.3, 0.26, 0.16 (cm) respectively.
  • Prohep treatment reduced the mean maximum tumor size by 70.51%compared with the control. While the old formula of Prohep showed reduction of the tumor size by 40%compared with the control.
  • Prohep In addition to reducing the HCC incidence, Prohep also impeded the progression of MASLD in DEN-HFD HCC mice model.
  • the anti-tumor effects were accompanied by ameliorated MASH progression, as evidenced by lower hepatic steatosis, fibrosis area, and NAS score (Fig. 2) .
  • Hepatic inflammation, such as macrophage infiltration revealed by F4/80 IHC staining (Fig. 2) and reduced pro-inflammatory cytokines TNF- ⁇ , IL-17, IL-1 ⁇ IL-6 were also ameliorated in the Prohep-treated group.
  • the improvement of liver health was secondly shown in reduced serum levels of ALT, AST, cholesterol and leptin.
  • Low dose Sorafenib treatment ameliorated the major hallmarks of MASLD including hepatic steatosis, inflammation and fibrosis but liver markers such as cholesterol, ALT and AST, and leptin in serum.
  • Pro-Sora and PrePro-Sora have significantly reduced TNF- ⁇ level and less macrophage infiltration as indicated by F4/80 staining that could lead to NAS score decrease when compared to Sora group. Beyond that, PrePro-Sora could reduce the serum leptin level compared to Sora group, indicating a remission from leptin resistance.
  • Sora group also showed significantly reduced FAS, ACC1 expression accompanied with decreased liver total cholesterol and triglyceride levels when compared to Con group. It could be linked to the body and liver weight drop after Sorafenib treatment. Compared to Sora group, PrePro-Sora showed a significantly increased hepatic SOD level that could help reduce the oxidative stress in the tumor microenvironment.
  • PI3K/mTOR pathway is responsible for downstream signals that promote cancer cell growth and proliferation, making it a crucial target for HCC treatment.
  • the protein expression of phosphorylated PI3K and mTOR were significantly reduced, leading to inhibited cancer proliferation signaling (Fig. 4) .
  • phosphorylated PI3K level was significantly suppressed in PrePro group that could contribute to anti-tumor effect.
  • the gut microbiota compositions of all groups were investigated. Notably altered microbial composition was observed in Sora, Pro, Pro+Sora, PrePro+Sora compared to Con group as illustrated in the PCoA plot (Table 1) . Notably, PrePro-Sora group showed significant differentiation from Sora group in gut microbiota composition while Pro-Sora group did not (Table 1) , indicating the modulation effects of giving Prohep in advance.
  • the analysis of alpha diversity did not reveal any notable differences among the groups studied. This observation is consistent with the results of a recent study of MSALD-HCC gut microbiota. It found that HFD feeding significantly reduced alpha diversity after 2 weeks compared to normal chow feeding. However, there was no significant difference in alpha diversity between the HFD and normal chow feeding groups after a longer period of 16 weeks or 14 months.
  • the metagenomic predicted functions were in consistency with the increased fecal propionic acid concentration in Pro group (Fig. 5D) .
  • PrePro-Sora 854 correlations
  • Sora 720 correlations
  • Pro-Sora 638 correlations
  • PrePro-Sora (296 negative correlations) and Pro-Sora (206 negative correlations) groups had more negative correlations compared to Sora (150 negative correlations) .
  • probiotics species contributed to the inhibition of certain known pathogenic species, say, B. breve, L. acidophilus, L. paracasei, L. rhamnosus, L. plantarum, L. acidophilus, S. thermophilus against Bacteroides faecium.
  • butyric acid were promoted in both Pro-Sora and PrePro-Sora groups. Increased butyric acid was positively correlated with the abundance of Brachyspira murdochii, Brachyspira hydodysenteriae, Brachyspira hampsonii, L. helveticus, L. acidophilus, B. breve, B. animalis ssp. lactis.
  • thermophilus were identified to be negatively correlated with tumor counts and tumor maximum size again in the correlation analysis includes only Sora, Pro-Sora, and PrePro-Sora group. tumor counts and tumor maximum size were negatively correlated with Helicobacter typhlonius and Helicobacter apodemus as well.
  • gut microbiota has been increasingly recognized.
  • Several studies have highlighted the occurrence of gut dysbiosis in various stages of chronic liver diseases, ranging from MASLD to cirrhosis and HCC. Additionally, gut microbiota has implications for the pharmacokinetics of cancer drugs and the management of cancer treatment.
  • the anti-tumorigenic effect of Prohep was demonstrated and the adjuvant effect of Prohep to low dose-Sorafenib treatment in DEN-HFD induced MASLD-HCC mouse model.
  • the current study was the first to explore the adjuvant effect of probiotics on low dose Sorafenib, which considers both the treatment efficacy and side effect management.
  • M. intestinale was found to be enriched in a blueberry malvidin-3-galactoside-treated HepG2 cell-injected tumor model and identified as an important factor affecting the microbial tricarboxylic acid (TCA) cycle KEGG pathway. Additionally, the significantly elevated species P. vulgatus was shown to mitigate the development of MASLD by promoting the production of metabolite 3-Hydroxyphenylacetic acid (3-HPAA) , which restrained lipid accumulation in liver cells. Furthermore, altered gut microbiota was found to antagonize pathogenic bacteria in MASLD-HCC, with the growth of pathogenic species such as T.
  • TCA microbial tricarboxylic acid
  • CRC colorectal cancer
  • E. lenta has been reported to be enriched in multiple acute and chronic inflammatory diseases.
  • F. rodentium originated from genus Faecalibaculum, is well studied for being proinflammatory bacteria that may impair the gut barrier as well.
  • SCFA short-chain fatty acid
  • HDAC histone deacetylases
  • Modulating the tumor microenvironment is a strategy for targeting HCC.
  • Propionate has previously been shown to regulate hepatic glucose and lipid metabolism via AMPK activation in human HepG2 hepatocytes.
  • liver AMPK was activated in the Prohep group.
  • increased propionate level has been linked to the activation of liver AMPK.
  • Liver cancers are characterized by upregulation of lipid catabolism and oxidative stress.
  • AMPK as a primary regulator of cellular energy homeostasis, often contributes to metabolic-associated disorders and is commonly found to be dysfunctional in patients with HCC. Low AMPK activation is correlated with aggressive clinicopathologic features and poor prognosis.
  • activated AMPK can inhibit the synthesis of fatty acids and cholesterol by downregulating the expression of lipogenesis genes such as FAS and ACC.
  • AMPK activation has also been reported as an inducer of CD36 expression, leading to lipid accumulation in hepatocytes.
  • AMPK activation decreased expression of FAS, ACC, and CD36 was observed in the Prohep group.
  • Activation of the AMPK signaling pathway has been shown to contribute to the regulation of reactive oxygen species (ROS) .
  • ROS reactive oxygen species
  • AMPK regulates FOXO3A and HO-1, downstream target genes, in response to cellular oxidative stress. Both FOXO3A and HO-1 contribute to the prevention of the increase of oxidative stress.
  • Metformin supplementation has been reported to inhibit the development of hepatocellular carcinoma via FOXO3 activation through the AMPK pathway. Furthermore, increased SOD and GSH activity helped to relieve oxidative stress due to hepatic fatty acid overload. Overall, the decrease in fatty acid uptake, synthesis of lipids and oxidative stress damage hindered the lipotoxicity-driven pathogenesis of metabolic diseases and lowered the risk of HCC.
  • the pro-inflammatory and pro-fibrotic liver microenvironment promotes hepatocarcinogenesis.
  • inflammation in the liver as represented by the macrophage marker F4/80 and expression levels of TNF- ⁇ , IL-17, IL-6, and IL-1 ⁇ , were significantly reduced.
  • fibrosis scars were also reduced after Prohep supplementation.
  • the multiple-hit hypothesis of the pathogenesis of MASH and subsequent HCC development suggests that multiple hepatotoxic insults occurring in parallel contribute to disease progression.
  • Oncogenic signaling transduction pathways such as the phosphoinositide 3-kinase (PI3K) , AKT, and mammalian target of rapamycin (mTOR) pathways, which play a major role in regulating cell proliferation, survival, and angiogenesis, are of great importance in cancer therapy.
  • Dysregulation of the PI3K/AKT/mTOR signaling pathway is common in HCC.
  • Previous studies have demonstrated that sodium propionate supplementation suppressed the PI3K/Akt/mTOR signaling pathway and attenuated LPS-induced epithelial-mesenchymal transition. In the current study, PI3K/mTOR signaling was suppressed in the Prohep group, leading to inhibited cancer cell proliferation.
  • Prohep supplementation effectively inhibits the development of MASLD-HCC, as demonstrated by a reduced tumor count, and maximum tumor size compared to the control group.
  • Proheps were found to alleviate crucial factors of MSALD-HCC development, such as steatosis, fibrosis, and inflammation.
  • Ki67 A sharp decrease in the proliferation marker Ki67 was also observed across all treatment groups.
  • Prohep supplementation led to the activation of AMPK, which plays an important role in cellular and systemic energy homeostasis.
  • Prohep group inhibited PI3K/mTOR pathway, a cancer proliferation pathway.
  • the therapeutic effect of Prohep can be partially attributed to the modulation of gut microbiota and the increased production of propionate and valerate.
  • Additional supplementation of Prohep to low-dose Sorafenib significantly reduced tumor counts in MASLD-HCC model while low-dose sorafenib alone didn’ t.
  • the potentiating effects could be attributed partially to ameliorated liver inflammation and increased antioxidative activity.
  • the current findings demonstrate the prophylactic potential of Prohep alone and adjuvant effect on low dose sorafenib.
  • Example 2 Prohep attenuates colorectal tumorigenesis in murine AOM/DSS model by suppressing STAT3, inducing apoptotic p53 and modulating gut microbiota
  • Azoxymethane (AOM) and dextran sulfate sodium (DSS) were purchased from Sigma-Aldrich (St. Louis, MO, USA) and TdB Labs (Uppsala, Sweden) respectively.
  • Prohep probiotic mixture formula composed of Lactobacillus helveticus, Bifidobacterium lactis, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus paracasei , Lactobacillus acidophilus, Bifidobacterium breve and Streptococcus thermophilus.
  • Prohep was produced in lyophilized powder under GMP (Fukopharma, Finland) . All primary antibodies were bought from Abcam (Cambridge, UK) and CST (Massachusetts, USA) while the secondary antibodies were purchased from Bio-Rad (Hercules, CA, USA) .
  • mice were obtained from the Centre for Comparative Medicine Research (CCMR, HKU) . 6 weeks old male BALB/c mice were placed in 12-hour light/dark cycle with a normal chow diet and drinking water given ad libitum for acclimatization upon the first week of receival. Voluntary gel administration training was carried out for the week afterwards. The chow diet being was taken away overnight on the first day and 0.20 g of Sucralose gel (Maine, USA) was given the next morning in individual cages. Mice were allowed to return to their house cages after consuming the given gel. The training continued for another two days but chow diet intake was not constrained. Mice group allocation was as follows: 1. Healthy control (H) , 2.
  • AOM/DSS control (AOMDSS) , 3.5-FU (F) , 4. Prohep (P) and 5. Prohep+5-FU (PF) .
  • AOM/DSS model was established by injecting the mice with AOM (10mg/kg) intraperitoneally at the start of the experiment while PBS was injected for the healthy control group.
  • One week of DSS (2.5%) , added in drinking water, was provided and be replaced by regular drink water in the next week. Three cycles of DSS treatment were given in total. Healthy control group received regular drinking water for all time. Prohep administration started at week 0.
  • Prohep (7 ⁇ 10 9 CFU per mice) were infused into Sucralose gel (Maine, USA) and provided to each mouse of Prohep and Prohep+5-FU groups at 0.20 g every other day until the end of the experiment.
  • the dosage of Prohep was based on the previous studies.
  • the other groups received Sucralose gel without modification as control.
  • 5-FU 35 mg/kg
  • PBS vehicle
  • the animals were sacrificed, and the colon was cut open longitudinally to determine the tumor count and colon length.
  • Colon samples were frozen at -80°C for biochemical analysis and were fixed in 10%formalin for histology analysis. Fecal samples were collected and frozen at -80°C on the sacrifice day. Liver samples were weighted. All protocols and procedures were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong (CULATR No. 5584-20) .
  • H&E staining was performed according to the manufacturer’s manual (BASO, Wuhan, China) . Adenoma counts were performed with light microscopy. Sirius red staining was carried out according to the manufacturer’s manual (Abcam, Cambridge, UK) . The percentage of positive red staining area over the total colon area measured was used to quantify the extent of colonic collagen fibrosis.
  • IHC staining was performed with heat-induced antigen retrieval method with sodium citrate (pH6) or Tris-EDTA (pH9) . Endogenous peroxidase activity was inhibited with 3%H 2 O 2 and sections were blocked with CAS-block reagent (Invitrogen, Waltham, MA, USA) for one hour. Endogenous peroxidase activity was inhibited with 3%H 2 O 2 and blocked with CAS-block reagent (Invitrogen, Waltham, MA, USA) for one hour. Primary antibodies incubation (1: 100) was performed at 4°C overnight followed by secondary antibodies incubation (1: 250) at room temperature for one hour.
  • DAB (Abcam, Cambridge, UK) chromogen reaction and haematoxylin counterstaining were performed to visualize the positively stained area.
  • the percentage of positively stained area was determined with ImageJ software (NIH, USA) and histoscore was calculated by multiplying the percentage with the intensity of staining graded from 0, non-stained; 1, weakly stained; 2, moderately stained; and 3, strongly stained.
  • Protein extraction was performed by homogenizing colonic samples in RIPA buffer with protease and phosphatase inhibitor (Sigma-Aldrich, St. Louis, MO, USA) and protein samples were collected by centrifuging the homogenate. The total protein content was measured using DC protein assay (Bio-Rad, CA, USA) .
  • ELISA analysis of TNF- ⁇ was performed using Mouse ELISA MAX TM Set (BioLegend, CA, USA) according to the instructions of manufacturer. The absorbance was measured using SpectraMax iD3 microplate readers (Molecular devices, CA, USA) .
  • Diluted protein was electrophorized in 10%SDS-PAGE gel and transferred to a polyvinylidene fluoride (PVDF) membrane.
  • the membrane was blocked with 5%non-fat milk or BSA, followed by overnight exposure of anti-p53 (1: 1000) (Abcam, Cambridge, UK) at 4°C. Secondary antibody exposure was performed the next day with goat anti-rabbit IgG (H+L) HRP conjugate or goat anti-mouse IgG (H+L) HRP conjugate (1: 4000, Bio-Rad, CA, USA) for one hour at room temperature. Protein bands were visualized with enhanced chemiluminescence reagents (Bio-Rad, CA, USA) using ChemiDox XRS+ imaging system (Bio-Rad, CA, USA) .
  • Fecal samples obtained on the sacrifice date were used to extract microbial DNA using Pro DNA kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. Extracted DNA was sent to BGI Genomics (Shenzhen, China) for whole genome shotgun sequencing. SOAPnuke was used to filter out adaptor reads that were of low quality or artificial. Bowtie2 was used to exclude the reads mapped to phix and mice contamination (GRCm39) genomes. Using the most recent release (2023-05-10) of the NCBI nr database, which contains archaea, bacteria, viruses, fungi, and microbial eukaryotes, taxonomic profiles were produced using Kaiju with parameter "-e 5" .
  • Taxa excluded from the downstream analysis were those that were either present in less than 10%of the samples or had a relative abundance of less than 0.01%.
  • R package phyloseq was used to aggregate the raw abundance table into a species-level counts per million (CPM) table.
  • CPM species-level counts per million
  • R package vegan was then used to determine alpha and beta diversity.
  • Dunn test and PERMANOVA using adonis2 from the R package vegan) were applied for alpha diversity and beta diversity, respectively, to assess the overall composition difference between groups.
  • ANCOM-BC was used to compare the abundance of microorganisms between AOM/DSS with and without treatment.
  • Linear discriminant analysis (LDA) was also used to lessen the bias present in the various differential methods.
  • Microorganisms that met certain criteria were deemed differentially abundant, including Fold change ⁇ 2 or ⁇ 1/2, LDA score (1og10 transformed) ⁇ 2, and FDR ⁇ 0.05.
  • Concatenated paired-end read files were input to HUMAnN3 (v3.7) in order to evaluate the abundance of metabolic Metacyc pathways and gene families abundance.
  • Kyoto Encyclopedia of Genes and Genomes (KEGG) ontology (KO) , KEGG module, and Metacyc pathway profiles were obtained by utilizing the HUMAnN3 regroup table function.
  • ANCOM-BC was used to assess the differential abundances (DA) of KO and the abundance of metabolic pathways. KO and pathways that met FDR value ⁇ 0.1 were deemed differentially abundant.
  • Fecal SCFAs content was determined using gas chromatography-mass spectrometry (GC-MS) according to previous studies.
  • fecal samples were homogenized in in 0.005 M sodium hydroxide with internal standard (10 ⁇ g/mL acetic acid-d4) and centrifuged at 13, 200 ⁇ g for 20 mins. The supernatant was then mixed with 0.5mL of 1-propanol/pyridine (3: 2, v/v) and 0.1mL of propyl chloroformate.
  • the SCFAs were derivatized by vertexing the mixture for 1 minute and incubating it at 60°C. 0.5mL hexane was then added, vortexed, and centrifuged at 2000 ⁇ g for 5 minutes.
  • Prohep as the only treatment group to reduce the degree of collagen fibrosis in colon.
  • the proliferative and apoptotic status was evaluated with IHC staining.
  • Prohep and Prohep+5-FU significantly reduced the H-score of the proliferative marker, Ki67 (p ⁇ 0.05) , but 5-FU did not (Fig. 8B, Fig. 8I) .
  • 5-FU and Prohep improved the H-score of the apoptotic marker, c-casp3 (p ⁇ 0.05) , but not by Prohep+5-FU (Fig. 8F, Fig. 8I) .
  • AOM/DSS elevated the levels of pro-inflammatory cytokine, TNF- ⁇ (p ⁇ 0.05) (Fig. 8A) .
  • All treatments including 5-FU, Prohep and Prohep+5-FU significantly reduced the concentration of TNF- ⁇ (p ⁇ 0.05) (Fig. 8A) .
  • No suppressive effects on cancer cell progression were observed in the 5-FU and Prohep+5-FU groups.
  • 5-FU also demonstrated elevation in the apoptotic marker, c-casp3 (p ⁇ 0.05) (Fig. 8F, Fig.
  • Prohep significantly modulated the abundance of gut microbiota compared to AOM/DSS.
  • Prohep elevated the abundance of Ligilactobacillus ruminis, Ligilactobacillus murinus, Adlercreutzia caecimuris, Ligilactobacillus animalis, Adlercreutzia mucosicola, Enterococcus faecalis, Sangeribacter muris, Muribaculaceae bacterium Isolate-001 (NCI) , Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila, Muribaculaceae bacterium Isolate-080 (Janvier) , Duncaiella dubosii, Prevotella sp.
  • PTAC Muribaculaceae bacterium Isolate-007
  • NCI Muribaculaceae bacterium Isolate-007
  • Hcobacter typhlonius p ⁇ 0.05
  • Prohep lowered Clostridum sp.
  • CAG 510, bacterium 0.1xD8-71, Candidatus Gastranaerophilus sp. (ex Termes propinquus) , Vermiculatibacterium agrestimuris, Roseburia sp.
  • CAG 309 (p ⁇ 0.05) (Fig. 9A) .
  • Helicobacter ganmani, Helicobacter hepaticus and Candidatus Borkfalkia ceftriaxoniphila were also inversely correlated to tumor count ⁇ 2mm (p ⁇ 0.05)
  • Desulfovibrio porci and Candidatus Borkfalkia ceftriaxoniphila were inversely correlated to tumor count ⁇ 2mm (p ⁇ 0.05) (Fig. 9B) .
  • a correlation analysis between the enriched species was carried out to study the possible role of bacteria among the microbiota community of AOM/DSS and Prohep.
  • Desulfovibrio porci Prevotella sp.
  • PTAC Helicobacter ganmani and Muribaculaceae bacterium Isolate-007
  • NCI Muribaculaceae bacterium Isolate-007
  • PTAC demonstrated the highest number of correlations with other bacteria and negatively correlated with most of the AOM/DSS enriched bacteria and positively correlated with other Prohep enriched bacteria.
  • Prevotella sp. PTAC and Desulfovibrio porci were significantly correlated with each other (p ⁇ 0.05) (Fig. 10) .
  • the enriched Desulfovibrio porci was the only species found to be negatively correlated with all AOM/DSS enriched species and total tumor count at the same, highlighting its importance in attenuating CRC gut dysbiosis and tumorigenesis.
  • Prohep On governing metabolic and biosynthesis pathways in comparison with AOM/DSS were studied from Metacyc and KEGG analysis.
  • Metacyc Prohep downregulated 17 pathways and upregulated 26 pathways (Fig. 11A) .
  • Prohep reduced peptidoglycan biosynthesis II staphylococci
  • dTDP-3-acetamido- ⁇ -D-fucos biosynthesis p ⁇ 0.05
  • LPS lipopolysaccharide
  • Prohep also downregulated purine nucleotides degradation II (aerobic) (p ⁇ 0.05) , which limits the conversion of purine into cancer related uric acid.
  • Prohep elevated pathways related to the biosynthesis of beneficial compounds, including L-lysine biosynthesis I (p ⁇ 0.05) and L-lysine biosynthesis II (p ⁇ 0.05) which were related to the biosynthesis of L-lysine; octanoyl- [acyl-carrier protein] biosynthesis (mitochondria, yeast) (p ⁇ 0.05) for the biosynthesis of lipoic acid; pyrimidine deoxyribonucleotides biosynthesis from CTP (p ⁇ 0.05) , superpathway of pyrimidine deoxyribonucleotides de novo biosynthesis (p ⁇ 0.05) and superpathway of pyrimidine ribonucleotides de novo biosynthesis (p ⁇ 0.05) for the biosynthesis of pyrimidine; Rubisco s
  • Prohep also upregulated pathways energy utilization pathway of lactic-acid producing bacteria, lactose and galactose degradation I (p ⁇ 0.05) and TCA cycle VII (acetate-producers) (p ⁇ 0.05) which indicates for greater abundance of acetic acid producing bacteria.
  • Prohep downregulated 23 KO gene and upregulated 55KO gene (Fig. 11B) .
  • Prohep increased aminodeoxyfutalosine deaminase (p ⁇ 0.05) , aminodeoxyfutalosine synthase (p ⁇ 0.05) , 1, 4-dihydroxy-6-naphthoate synthase (p ⁇ 0.05) , chorismate dehydratase (p ⁇ 0.05) , cyclic dehypoxanthinyl futalosine synthase (p ⁇ 0.05) , adenosylhomocysteine/aminodeoxyfutalosine nucleosidase (p ⁇ 0.05) , acetyl-CoA acyltransferase (p ⁇ 0.05) , hydroxymethylbilane synthase (p ⁇ 0.05) , porphobilinogen synthase (p ⁇ 0.05) , glutamate-1-semialdehyde 2, 1-aminomutase (p ⁇ 0.05)
  • the fecal SCFA concentration was accessed based on the results of Metacyc analysis. In comparison with the healthy group, the fecal acetate concentration was lowered in the AOM/DSS, 5-FU and Prohep +5-FU groups (p ⁇ 0.05) (Fig. 9C) . Meanwhile, the fecal acetate concentration in Prohep was significantly increased compared to the AOM/DSS group (p ⁇ 0.05) . Furthermore, the relative abundance of acetate-producing Acidiphilium genus and Acidiphilium sp. CAG: 727 were upregulated by Prohep in comparison to AOM/DSS (p ⁇ 0.05) .
  • Prohep significantly alleviated AOM/DSS-induced colorectal tumorigenesis by suppressing proliferative STAT3, inducing apoptotic p53, modulating the gut microbiota, and elevating acetate concentration.
  • Prohep also demonstrated the strongest alleviative effects compared to 5-FU, and Prohep+5-FU.
  • all experimental mice developed colorectal tumors from the AOM/DSS treatment.
  • some mice in AOM/DSS, 5-FU, and Prohep+5-FU groups reached humane endpoints that required euthanasia as determined by animal facility veterinarians.
  • only Prohep significantly improved the survivability of experimental animals, but not in 5-FU and Prohep+5-FU groups.
  • Prohep demonstrated anti-inflammatory responses in the colon. Inflammation-related histopathological parameters, including hyperplasia, disrupted colonic structure, lengthened colonic crypt depth, and collagen fibrosis, were all alleviated by Prohep.
  • the pro-inflammatory cytokines TNF- ⁇ and inflammatory regulator STAT3 activation were suppressed.
  • TNF- ⁇ was found to promote proliferation and inhibit apoptosis in colon cancer cells by activating STAT3.
  • STAT3 was suggested to be a mediator connecting inflammation and cancer progression, in which silencing of the STAT3 was found to limit CRC cell growth and induce cell death at the G2/M stage.
  • Prohep also downregulated c-jun and c-fos expression levels which functioned as the effectors of STAT3-induced tumor cell proliferation. Furthermore, STAT3 silencing also led to apoptosis through upregulation of p53 and caspase-3. Similar results were observed in the present study with the elevated expression level of p53 and c-casp3.
  • p53 serves an important function in suppressing tumor growth by inducing cell cycle arrest, DNA repair, senescence and apoptosis. Deficiencies of p53 were often found in cancer and the restoration of p53 was targeted for cancer therapy including CRC. At the same time, p53 also inhibits STAT3 activators and attenuates STAT3 functions. Given both STAT3 and p53 are effective targets and the presence of opposing STAT3-p53 regulatory loops, co-targeting STAT3 and p53 by Prohep was therefore suggested to be promising in CRC regulation.
  • Prohep administration also altered the gut microbiota.
  • Prohep enriched the relative abundance of Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila and Helicobacter typhlonius, which showed a reverse correlation with colorectal tumor counts in this study.
  • Helicobacter ganmani and Helicobacter hepaticus are two Helicobacter species naturally colonizing mouse gut which are reported to be related to IBD development.
  • Desulfovibrio porci was also found to be inversely correlated to tumor count. Except for identifying it as a hydrogen sulfide (H 2 S) producing species, its effects on animals were not well-documented. H 2 S was reported with dual effects on cancer cells following a biphasic dose-response curve. Recent studies suggest that H 2 S exposure in high concentrations for prolonged periods induced apoptotic responses in cancer cells both in vivo and in vitro. Thus, it was proposed that the H 2 S-producing properties of Desulfovibrio porci might contribute to the alleviation of colorectal tumorigenesis in this study which is worth further validation in the future. Furthermore, the enriched Candidatus Borkfalkia ceftriaxoniphila, a newly discovered and low abundant commensal species, had a significant increase in abundancies accompanying the growth of probiotics in the human gut.
  • the inner correlation analysis identified Prohep-enriched Desulfovibrio porci, Prevotella sp. PTAC, Helicobacter ganmani and Muribaculaceae bacterium Isolate-007 (NCI) were inversely correlated to AOM/DSS enriched bacteria. Both Desulfovibrio porci and Prevotella sp. PTAC displayed a significant role in Prohep-induced gut microbiota modulation and a positive correlation in relative abundance was found between them. A higher abundance of Prevotella was reported to be linked to lower risks of CRC progression in CRC patients. Although Prevotella sp.
  • PTAC was not found to be significantly correlated to reduced tumor count in this study, its co-occurrence with Desulfovibrio porci and other Prohep-enriched bacteria suggested it played a vast role in supporting the growth of these beneficial bacteria in CRC management. Notably, only the enrichment of Desulfovibrio porci negatively correlated to all AOM/DSS enriched bacteria. Given its negative correlation with CRC tumor count as well, the upregulation of Desulfovibrio porci was suggested to be crucial in alleviating CRC tumorigenesis through reversing AOM/DSS-induced gut microbiota.
  • Prohep Apart from the correlation against the colorectal tumorigenesis-related parameters, the mechanisms behind the anti-CRC effects of Prohep was also studied through the prediction of the metabolic functions from Metacyc and KEGG analysis. From the Metacyc analysis, Prohep downregulated the CRC-related peptidoglycan and LPS biosynthesis as well as the conversion of uric acid from purine. Peptidoglycan is a major component of the gram-positive bacteria cell wall. When peptidoglycan is transferred across the intestinal epithelial cells, it interacts with macrophages, triggers IL-6 secretion and induces inflammation and fibrosis in Crohn’s disease and CRC.
  • LPS is a cellular component from bacterial membranes which is known for its endotoxic properties. LPS would induce inflammation by activating nuclear factor- ⁇ B (NF- ⁇ B) pathway via Toll-like receptor 4 (TLR4) which exacerbates gut barrier dysfunction and promotes CRC development. In addition, in the catabolism of purine nucleotides, uric acid is released which exhibits a positive association with CRC incidence .
  • NF- ⁇ B nuclear factor- ⁇ B pathway
  • TLR4 Toll-like receptor 4
  • L-lysine While L-lysine does not contribute directly to cancer management, it could be completely be converted into butyrate and acetate by gut microbiota like Intestinimonas strain AF211 and served a significant role in inhibiting CRC and maintaining the growth of normal colonocytes. Meanwhile, the combination of lysine with epigallocatechin gallate (EGCG) were found to enhance the inhibitory effects of EGCG on the growth of colon cancer cell line HCT 116.
  • EGCG epigallocatechin gallate
  • Alpha-lipoic acid was found to have anti-CRC by triggering cell death in tumor cells and could be combined with doxorubicin or 5-FU to synergistically kill CRC cells.
  • LA also prevent inflammatory and oxidative responses by suppressing the NF- ⁇ B pathway, as well as downregulating TNF- ⁇ , IL-6, COX-2, MDA, and MPO.
  • Pyrimidine and its derivatives were reported with pharmacological and anti-cancer properties, in which pyrimidine metabolic pathways were reported to regulate the chemotherapeutic effects of chemo drugs like 5-FU, tegafur and thioguanine.
  • N- [2- (dimethylamino) ethyl] -2, 3-dimethyl-4-oxo-4H-pyrido [1, 2-a] thieno [2, 3-d] pyrimidine-9-carboxamide (PTP) showed strong antitumor effects against human CRC cells by activating p53.
  • Palmitate is one of the common saturated fatty acids found in animals and plants which previous studies reported that a low intake of palmitic acid showed inverse associations with CRC and palmitic acid alongside ceramide were strong inhibitors of the EMT signaling axis of colorectal cancer cells.
  • Prohep elevated metabolic pathways related to energy utilization of lactic acid-producing bacteria (LAB) and acetate producers. Since Prohep is composed mostly of LAB, the increase of lactose and galactose degradation was expected and intestinal galactose was reported to have a protective effect against CRC by inhibiting mucosal proliferation. Besides, the TCA cycle of acetic acid bacteria was enhanced by Prohep. Acetic acid bacteria are known for their properties in oxidizing ethanol to acetic acid and the abundance of acetate-producing Acidiphilium was elevated. Acetate was identified as one of the important metabolites which inhibited CRC tumorigenesis in both in vivo and in vitro models.
  • Acetate reduced the tumor size of a CRC-cell-injected xenograft mouse model. It was suggested that acetate enhanced growth arrest and apoptosis of CRC cells through increasing oxygen consumption and reactive oxygen species production. Previous studies demonstrated more pronounced apoptotic effects on CRC cells compared to normal colonocytes, achieved by enhancing MCT1, MCT4 and CD147 while also re-localizing MCT1 at the plasma membrane. Also, acetate would serve as an energy substrate for normal colonocytes via de novo lipogenesis and allows the normal cells to outcompete cancer cells, which primarily rely on glycolysis for energy production. Given the elevation of pathways related to SCFAs and acetate production from Metacyc analysis, the fecal SCFAs profile was evaluated.
  • Arylformamidase (AFMID) was involved in the conversion of tryptophan into kynurenine by the transcription factor MYC in both cultured CRC cells and CRC patients. Phosphoserine aminotransferase is responsible for serine biosynthesis and an elevated serine was suggested to enhance the growth of CRC cells. The overexpression of phosphoserine aminotransferase was reported to heighten CRC cells chemoresistance and cell growth.
  • the serum activity of beta-glucuronidase was found to be higher in CRC patients than in healthy subjects, which suggested it as a marker of CRC.
  • alcohol dehydrogenase NADP+
  • H 2 O 2 hydrogen peroxide
  • Transketolase was abnormally increased in CRC which promoted cancer cell glycolysis by enhancing AKT phosphorylation and eventually worsening CRC metastasis.
  • Purine-nucleoside phosphorylase PNP was identified as a cancer marker as plasma PNP levels on average four times higher in cancer patients.
  • the expression level of PNP was also correlated with lymph vessel invasion, positive lymph node metastasis and advanced stage of CRC. The downregulation of these CRC-related pathways strengthened Prohep’s promising effects in inhibiting colorectal tumorigenesis.
  • Prohep elevated multiple beneficial pathways in the regulation of CRC.
  • Aminodeoxyfutalosine deaminase, aminodeoxyfutalosine synthase, 1, 4-dihydroxy-6-naphthoate synthase, chorismate dehydratase, cyclic dehypoxanthinyl futalosine synthase, and adenosylhomocysteine/aminodeoxyfutalosine nucleosidase are related to menaquinones which are the bacterial forms vitamin K produced by gut microbiota. Menaquinone or vitamin K2 was found to reduce KRAS proliferation in colon cancer cells and promote apoptotic cell death in CRC mice.
  • Acetyl-CoA acyltransferase was shown to have a negative correlation with the resistance of the targeted cancer drug, cetuximab, in CRC which the overexpression of ACAA would suppress proliferation and lower cetuximab tolerance in CRC cells.
  • Hydroxymethylbilane synthase was identified as a tumor suppressor gene and its inactivation was identified in patients with intermittent porphyria and sporadic HCC.
  • Porphobilinogen synthase is the crucial first step of tetrapyrrole biosynthesis which tetrapyrroles like unconjugated bilirubin, bilirubin ditaurate, biliverdin, biliverdin-/bilirubin dimethyl ester, urobilin, stercobilin and protoporphyrin exhibited DNA-damaging and apoptosis in colon and liver cancer cells. Meanwhile, porphobilinogen synthase, along with glutamate-1-semialdehyde 2, 1-aminomutase and glutamyl-tRNA reductase are involved in the production and utilization of aminolevulinic acid (ALA) which was shown to inhibit CRC cells.
  • ALA aminolevulinic acid
  • Fructose-1, 6-bisphosphatase I could inactive NF- ⁇ B which suppresses CRC and similarly, fructose-1, 6-bisphosphatase II was found to regulate gastric cancer in an inversed relationship.
  • 2-amino-4-hydroxy-6 hydroxymethyldihydropteridine diphosphokinase/dihydropteroate synthase was required in the biosynthesis of tetrahydrofolate which was associated with lowered risk of serrated polyps in CRC.
  • Prohep was found to be the most effective treatment for AOM/DSS mice when compared with 5-FU and Prohep+5-FU. No additive or synergistic effects were observed when Prohep was given in adjuvant with 5-FU. Prohep presented the strongest inhibition on colorectal tumorigenesis and inflammation among all groups. In the present study, even though 5-FU reduced the total tumor count in the colon, it failed to reduce the level of p-STAT3 and upregulate that of p53. The activation of STAT3 was reported to promote 5-FU resistance in CRC through increasing Mcl-1-dependent cytoprotective autophagy, by which 5-FU resistant cells would transfer p-STAT3-containing exosomes to the recipient cells and induce chemoresistance against 5-FU.
  • the loss or deficiency of p53 would contribute to 5-FU resistance and detriment the DNA damaging effects of 5-FU against CRC cells.
  • the restoration of p53 was reported to significantly improve 5-FU sensitivity in CRC cells. Nonetheless, even though Prohep increased p53 and reduced p-STAT3, the effects were not displayed when Prohep was co-treated with 5-FU. Besides a disrupted gut microbiome environment, 5-FU could lead to gut dysbiosis and intestinal mucositis. These effects may hinder the adhesion, colonization and activation of probiotic species and might thus limit the anti-CRC effects of Prohep when co-administrated.
  • Prohep reduced AOM/DSS-induced CRC carcinogenesis.
  • Prohep significantly reduced total tumor count, total tumor size, caecum weight, colonic crypt depth, colonic inflammation, and collagen fibrosis induced by AOM/DSS.
  • Prohep enriched the abundance of beneficial bacteria and acetate level which contributed to combat CRC.
  • Prohep showed superior anti-tumorigenesis effects compared to both 5-FU alone and Prohep+5-FU in the treatment of CRC.
  • Probiotic Mixture Attenuates Colorectal Tumorigenesis in Murine AOM/DSS Model by Suppressing STAT3, Inducing Apoptotic p53 and Modulating Gut Microbiota. Probiotics and Antimicrobial Proteins, 1-17.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

A probiotic composition for treating or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma(MASLD-HCC) or colorectal cancer, comprises one or more microbiota selected from a group consisting of Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus GG, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium animalis subsp. Lactis, Streptococcus thermophilus or any combination thereof. The probiotic composition demonstrates improved therapeutic effects when combined with low-dose Sorafenib for MASLD-HCC and outperforms 5-Fluorouracil in colorectal cancer models.

Description

PROBIOTICS MIXTURES AND METHODS OF USE THEREOF FOR TREATING CANCERS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims benefit of U.S. Provisional Application No. 63/555,341 filed Feb 19, 2024 and U.S. Provisional Application No. 63/555,348, filed Feb 19, 2024. Both applications 63/555,341 and 63/555,348 are hereby incorporated herein by reference in their entirety.
REFERENCE TO SEQUENCE LISTING
The Sequence Listing XML submitted as a file named “UHK_01473_PCT_ST26. xml, ” created on January 17, 2025, and having a size of 9, 889 bytes is hereby incorporated by reference pursuant to 37 C.F.R. § 1.834 (c) (1) .
FIELD OF THE INVENTION
The disclosed invention is generally in the field of cancer treatment and specifically in the area of treating cancers with probiotics.
BACKGROUND OF THE INVENTION
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent and multifaceted chronic hepatic disorder. It encompasses a range of pathological stages, including steatosis, metabolic dysfunction-associated steatohepatitis (MASH) , fibrosis, cirrhosis. MASLD elevates the risk of cardiovascular events, chronic kidney disease, as well as hepatic and extrahepatic malignancies, notably hepatocellular carcinoma (HCC) . The incidence of MASLD and its associated complications, particularly HCC, is anticipated to escalate in the foreseeable future. HCC is the second leading cause of cancer-related mortality globally. Nonetheless, no efficacious therapeutic interventions currently exist for MASLD-HCC.
A common communication exists between the gastrointestinal tract and the hepatic system, and is facilitated by the portal circulation, bile duct, and systemic circulation. The portal venous system supplies over two-thirds of the liver's blood from the intestinal tract, allowing intestinal bacteria and their constituents to efficiently reach the hepatic system. The translocated elements, such as bacterial products and lipopolysaccharides, can stimulate hepatic immune cells, activate inflammatory pathways, and ultimately contribute to the development of MASLD-HCC. These findings suggest the pivotal role of the gut-liver axis in the pathogenesis of MASLD-HCC and highlight a viable target for MASLD-HCC intervention.
The manipulation of gut microbiota using probiotics may offer a promising approach for liver diseases. Animal studies have demonstrated that probiotics can confer beneficial effects against MASLD. Moreover, numerous studies have reported the efficacy of probiotics as a prophylactic against different cancer types and as an adjuvant for cancer therapy to enhance its effectiveness. However, most studies have primarily investigated the early stages of MASLD, including liver steatosis and inflammation, while neglecting liver fibrosis, which is a hallmark of the advanced phase. Furthermore, the role of probiotics in MASLD-HCC remains poorly understood. Commensal probiotics from some genera such as lactobacillus, bifidobacterium and Butyricimonas in the gut were found to be diminished in MASLD-HCC patients.
Colorectal cancer (CRC) is the third most diagnosed cancer, as well as the second leading cause of cancer death worldwide. Over half of the cases are accounted for risk factors including smoking, an unhealthy diet, high alcohol consumption, physical inactivity, and excess body weight. The diagnosis of CRC is often at later stages, and its 5-year survival rate at the advanced stage is only 15%, indicating the importance of early diagnosis and effective treatment. Surgical treatments, including endoscopic mucosal resection and endoscopic submucosal dissection in the early stages and lymph node dissection in the advanced stages along with polyps removal, are the standard first-line treatment for CRC. Apart from surgical procedures, chemotherapeutic intervention is crucial to prevent and cure metastatic CRC and improve patients’ s urvival rates. Nonetheless, the response rate of the standard chemo drug backbone 5-fluorouracil (5-FU) on CRC is as low as 11%without combining with other regimens. Given the high economic burden of chemotherapy, affordable and effective pharmaceutical options are in demand to save lives.
An emerging direction for researching improved CRC treatments is microbiome-targeted therapy. Gut microbiota and cancer exhibit a bidirectional relationship, in which the alteration of microbial composition could promote pathogenesis while the modulation of gut microbiota could also be therapeutic for cancer. Gut microbiota’s influence on CRC initiation and progression was suggested to be both the “driver” and the “passenger” of the “driver/passenger” theory. The imbalance of the gut microbiota profile termed gut dysbiosis, was found to be a vital cause of colorectal tumorigenesis. On the contrary, modulation of the gut microbiota alleviated or prevented the progression of CRC in both healthy and cancerous subjects. Microbiome-targeted therapy in CRC treatment includes fecal microbiota transplant (FMT) , probiotics, diet and prebiotics, and antibiotic treatment. Unlike the complexity and high cost of personalized FMT, the damaging effects of antibiotics to commensal bacteria, the induction of probiotics in CRC treatment is a more affordable and efficient option for modulating the gut microbial composition. Evidence from studies has shown that probiotics are a successful treatment for colorectal cancer (CRC) by limiting the growth of pathogens, reversing dysbiosis, modulating the immune system, strengthening the intestinal barrier, and promoting an anti-cancer cell signaling network. 5-FU is an essential component in the standard chemotherapy of CRC and yet its efficacy was limited due to the development of chemoresistance and toxicity. Adjuvant combinations of probiotics with 5-FU were found to improve 5-FU applicability in CRC management. When treated with Lactobacillus and Bifidobacterium, the anti-cancer effects of 5-FU were enhanced with reduced intestinal toxicity and chemoresistance effects.
There is still a need for probiotic formulations that are beneficial for treating or preventing cancer.
It is an object of the present invention to provide composition and methods of treating a subject having cancer with a probiotic mixture.
Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present disclosure as it existed before the priority date of each claim of this application.
Throughout this specification the word “comprise, ” or variations such as “comprises” or “comprising, ” will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
BRIEF SUMMARY OF THE INVENTION
Described are compositions and methods for treating and/or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) using a probiotic formulation, Prohep. Also described are methods for treating and/or preventing cancer, particularly hepatocellular carcinoma and colorectal cancer using the probiotic formulation.
The probiotic formulation, Prohep, includes a mix of organisms selected from a group that includes Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus GG, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium animalis subsp. lactis and Streptococcus thermophilus.
In some forms, Prohep is formulated for oral administration. In some forms, Prohep compositions may also be administered orally, intracolonically, intranasally, intrarectally, via a catheter, via a lavage, via a nasogastric tube, via local delivery, or via a method for fecal microbiota transplantation (FMT) . In some forms, Prohep composition may be in the form of a dispersion. Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils.
In some forms, the current disclosure uses Prohep as an adjuvant therapy in combination with low-dose Sorafenib for the treatment of MASLD-HCC. In some forms, the Prohep can be used alone for the treatment of MASLD-HCC. In some forms, Prohep exhibits anti-tumorigenic properties by modulating the gut microbiota to increase the production of short-chain fatty acids (SCFAs) , including propionate, valerate, and butyrate, thereby improving gut dysbiosis and enhancing treatment efficacy. In some forms, the activation of AMPK by Prohep results in reduced lipogenesis, decreased lipid uptake, and upregulation of antioxidant enzyme expressions. In some forms, Prohep suppresses the PI3K/mTOR cancer proliferation pathway, contributing to reduced tumor counts, amelioration of inflammation, and increased hepatic superoxide dismutase (SOD) expression. In some forms, the combination of Prohep and low-dose Sorafenib leads to improved therapeutic outcomes without inducing adverse effects, as evidenced by decreased proliferation marker Ki67 expression, alleviation of hepatic inflammation, and maintenance of redox balance. The current disclosure highlights the modulation of Sorafenib-treated gut microbiota by Prohep, resulting in enhanced butyrate production and increased sensitivity to Sorafenib, establishing an important therapeutic approach for the neoadjuvant and long-term management of MASLD-HCC.
In some forms, the present disclosure uses Prohep for treating colorectal cancer. In some forms, Prohep demonstrates enhanced anti-tumorigenic effects compared to 5-Fluorouracil (5-FU) alone or in combination for treating colorectal cancer. In some forms, Prohep alleviates AOM/DSS-induced colorectal tumorigenesis by modulating inflammatory, proliferative, and apoptotic pathways, including downregulation of TNF-α and p-STAT3 and upregulation of p53. In some forms, Prohep reduces tumor burden, caecum weight, crypt depth, colonic inflammation, and collagen fibrosis induced by AOM/DSS. In some forms, the use of Prohep enriches beneficial bacteria such as Helicobacter ganmani, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila, Desulfovibrio porci, and Prevotella sp. PTAC, which were inversely correlated with tumor counts and suppressed AOM/DSS-associated pathogenic bacteria. In some forms, Prohep downregulated pathways associated with colorectal cancer (CRC) , including those involved in peptidoglycan, lipopolysaccharide (LPS) , and uric acid biosynthesis and conversion. In some forms, Prohep enhanced pathways related to CRC-suppressing metabolites such as menaquinone, tetrapyrrole, aminolevulinic acid, and tetrahydrofolate. In some forms, Prohep enhanced the biosynthesis of beneficial compounds like L-lysine, lipoic acid, pyrimidine, and palmitate and promoted metabolic pathways supporting energy utilization by lactic acid-producing bacteria (LAB) and acetate producers. In some forms, Prohep further upregulated fecal acetate concentration, contributing to its anti-CRC effects.
Additional advantages of the disclosed method and compositions will be set forth in part in the description which follows, and in part will be understood from the description, or can be learned by practice of the disclosed method and compositions. The advantages of the disclosed method and compositions will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed.
BRIEF DESCRIPTION OF THE DRAWINGS
The accompanying drawings illustrate several embodiments of the disclosed method and compositions and together with the description, serve to explain the principles of the disclosed method and compositions.
Figures 1A-1B show Prohep protects against NAFLD-HCC formation in DEN-HFD MASLD-HCC mouse models. (Fig. 1A) Experimental design for the dietary plan and treatment scheme in DEN+HFHC mouse model. (Fig. 1B) . Representative gross morphology, H&E-stained tumor area (T circled with red dotted line indicates tumor area) (100x) , IHC staining (200x) for Ki-67 and C-Casp3 in liver and H&E-stained (40x) histological images in lung (arrow indicates lung metastasis) of DEN+HFHC mouse model.
Figure 2 shows Prohep ameliorated MASLD progression in DEN-HFD HCC mice model. H&E (200x) , Oil-red O (200x) , Sirius red-stained (200x) non-tumor area and IHC staining (200x) for F4/80.
Figures 3A-3B show that Prohep modulated tumor microenvironment via activating AMPK signaling pathway its impact on cancer metabolism. (Fig. 3A) Representative IHC staining for AMPK images and scoring of liver tissues. (Fig. 3B) Protein expression of FAS, HO-1 and FOXO3A. Data are presented as the mean ± SD. *p < 0.05; **p < 0.01; ***p <0.001, ****p<0.0001.
Figure 4 shows that Prohep suppressed the activity of the PI3K/mTOR pathway in DEN-HFD induced HCC mouse models. Protein expression of P-PI3k, PI3K and mTOR. Data are presented as the mean ± SD. *p < 0.05.
Figures 5A-5D show that Prohep altered gut microbiota–related metabolites. (Fig. 5A) Significantly enriched KEGG pathways generated from fecal metagenomic data in Con and Pro groups. (Fig. 5B) Significantly altered enzymes participates the pathway “Pyruvate fermentation to propanoate I” . (Fig. 5C) Biosynthesis route of propionate. (Fig. 5D) Fecal SCFAs measurements.
Figures 6A-6F shows Prohep enhanced survivability and reduced colorectal tumorigenesis of AOM/DSS mice better than 5-FU and Prohep+5-FU. (Fig. 6A) Survival rate percentage. (Fig. 6B) Representative macroscopic pictures of the dissected colon. (Fig. 6C) Total tumor counts. (Fig. 6D) Total tumor size. (Fig. 6E) Caecum weight. (Fig. 6F) Liver weight. n = 6-10. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figures 7A-7F show that Prohep improved colorectal histopathology induced by AOM/DSS. (Fig. 7A) Colonic crypt depth. (Fig. 7B) Hyperplasia score. (Fig. 7C) Inflammation score. (Fig. 7D) Sirius red positive percentage. (Fig. 7E) Representative microscopic pictures of colonic sections under H&E staining. Colonic tumors were framed in rectangle and displayed in 100x. (Fig. 7F) Representative microscopic pictures of colonic sections under Sirius red staining. n = 6-10. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figures 8A-8I. Prohep suppressed pro-inflammatory and proliferative marker and promoted apoptotic markers. Colonic concentration of (Fig. 8A) TNF-α. Immunohistochemistry analysis H-score of (Fig. 8B) Ki67; (Fig. 8C) c-jun; (Fig. 8D) c-fos; (Fig. 8E) p-STAT3; and (Fig. 8F) c-casp3. Western blotting analysis of (Fig. 8G) p53. (Fig. 8H) Representative immunoblots of p53. (Fig. 8I) Representative microscopic pictures of colonic sections under IHC staining in 400x magnification. n = 6-10. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figure 9A-9C. Prohep modulated gut microbiota contributed to alleviate colorectal tumorigenesis and elevated SCFAs concentration. (Fig. 9A-9B) Heatmap showing the z-score transformed mean abundance of significantly altered species in H, F, P or PF as compared to AD (Fig. 9A) and Spearman’s correlation between relative bacterial abundance and fecal SCFA concentrations, colonic cytokines concentration, tumor counts and colorectal tumorigenesis related organs alterations (Fig. 9B) . (Fig. 9C) Fecal concentration of SCFA. AA: acetate; PA: propionate; BA: butyrate; IBA: isobutyrate; and VA: valerate. Differential abundant species based on ANCOM-BC and LDA score (FDR < 0.05, Fold change > 2 or < 1/2, LDA score (log10) > 2) were labelled with "*" in the right part. n = 6-10. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figure 10. Prohep enriched bacteria negatively correlated with AOM/DSS enriched bacteria. Inner correlation of enriched bacteria in AOM/DSS and Prohep group. Correlations with p-value < 0.05 and r > 0.3 were considered and visualized in the network. The green edge denotes for negative correlation and red edge denotes for positive correlation. The size of the node is proportional to the number of connections to other nodes.
Figure 11A-11B. Prohep modulated gut microbiota to induce anti-tumor metabolic functions. (Fig. 11A) Heatmap showing the z-score transformed mean abundance of KEGG orthology (KO) originated from three significantly altered KEGG modules in H, F, P or PF as compared to AD (left) . Differential KO based on ANCOM-BC (adjusted p-value < 0.1) were displayed in the right part; and (Fig. 11B) Significantly altered Metacyc pathway in H, F, T or TF as compared to AD. The color is proportional to log2 transformed fold change (Treatment/Control) , while the size is proportional to the value of -log10 transformed adjusted p (FDR-corrected) . n = 6-10. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figure 12: Diagram summarizing the proposed mechanism of the disclosed probiotics mixture.
DETAILED DESCRIPTION OF THE INVENTION
The disclosed method and compositions can be understood more readily by reference to the following detailed description of particular embodiments and the Example included therein and to the Figures and their previous and following description.
It is to be understood that the disclosed method and compositions are not limited to specific synthetic methods, specific analytical techniques, or to particular reagents unless otherwise specified, and, as such, can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
A. Definition
Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein.
Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains.
As used herein, the singular forms “a” , “an” , and “the” include both singular and plural referents unless the context clearly dictates otherwise.
Use of the term “about” is intended to describe values either above or below the stated value in a range of approx. +/-10%; in other forms the values can range in value either above or below the stated value in a range of approx. +/-5%; in other forms the values can range in value either above or below the stated value in a range of approx. +/-2%; in other forms the values can range in value either above or below the stated value in a range of approx. +/-1%. The preceding ranges are intended to be made clear by context, and no further limitation is implied.
As used herein, the term “therapeutic agent” refers to any agent that, when administered to a subject, has a therapeutic, prophylactic, and/or diagnostic effect and/or elicits a desired biological and/or pharmacological effect.
The term “modulate” as used herein refers to the ability of a compound to change an activity in some measurable way as compared to an appropriate control. As a result of the presence of compounds in the assays, activities can increase or decrease as compared to controls in the absence of these compounds. Preferably, an increase in activity is at least 25%, more preferably at least 50%, most preferably at least 100%compared to the level of activity in the absence of the compound. Similarly, a decrease in activity is preferably at least 25%, more preferably at least 50%, most preferably at least 100%compared to the level of activity in the absence of the compound. A compound that increases a known activity is an “agonist” . One that decreases, or prevents, a known activity is an “antagonist” .
The term “inhibit” means to reduce or decrease in activity or expression. This can be a complete inhibition or activity or expression, or a partial inhibition. Inhibition can be compared to a control or to a standard level. Inhibition can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100%. In some forms, the inhibition and reduction are compared according to the level of mRNAs, proteins, cells, tissues, and organs.
The term “monitoring” as used herein refers to any method in the art by which an activity can be measured.
The term “providing” as used herein refers to any means of adding a compound or molecule to something known in the art. Examples of providing can include the use of pipettes, pipettemen, syringes, needles, tubing, guns, etc. This can be manual or automated. It can include transfection by any mean or any other means of providing nucleic acids to dishes, cells, tissue, cell-free systems and can be in vitro or in vivo.
The term “preventing” as used herein refers to administering a compound prior to the onset of clinical symptoms of a disease or conditions so as to prevent a physical manifestation of aberrations associated with the disease or condition.
The term “in need of treatment” as used herein refers to a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals) that a subject requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a care giver's expertise, but that include the knowledge that the subject is ill, or will be ill, as the result of a condition that is treatable by the disclosed compounds.
As used herein, “subject” includes, but is not limited to, animals, plants, parasites and any other organism or entity. The subject can be a vertebrate, more specifically a mammal (e.g., a human, horse, pig, rabbit, dog, sheep, goat, non-human primate, cow, cat, guinea pig or rodent) , a fish, a bird or a reptile or an amphibian. The subject can be an invertebrate, more specifically an arthropod (e.g., insects and crustaceans) . The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. A patient refers to a subject afflicted with a disease or disorder. The term “patient” includes human and veterinary subjects. In some forms, the subject can be any organism in which the disclosed method can be used to genetically modify the organism or cells of the organism.
The terms “individual” , “host” , “subject” , and “patient” are used interchangeably, and refer to a mammal, including, but not limited to, murids, simians, humans, mammalian farm animals and livestock, mammalian sport animals, and mammalian pets.
“Treatment” or “treating” means to administer a composition to a subject or a system with an undesired condition (e.g., HCC) . The condition can include one or more symptoms of a disease, pathological state, or disorder. Treatment includes medical management of a subject with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological state, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological state, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological state, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological state, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological state, or disorder. It is understood that treatment, while intended to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder, need not actually result in the cure, amelioration, stabilization or prevention. The effects of treatment can be measured or assessed as described herein and as known in the art as is suitable for the disease, pathological condition, or disorder involved. Such measurements and assessments can be made in qualitative and/or quantitative terms. Thus, for example, characteristics or features of a disease, pathological condition, or disorder and/or symptoms of a disease, pathological condition, or disorder can be reduced to any effect or to any amount. “Prevention” or “preventing” means to administer a composition to a subject or a system at risk for an undesired condition. The condition can include one or more symptoms of a disease, pathological state, or disorder. The condition can also be a predisposition to the disease, pathological state, or disorder. The effect of the administration of the composition to the subject can be the cessation of a particular symptom of a condition, a reduction or prevention of the symptoms of a condition, a reduction in the severity of the condition, the complete ablation of the condition, a stabilization or delay of the development or progression of a particular event or characteristic, or reduction of the chances that a particular event or characteristic will occur.
As used herein, the terms “effective amount” or “therapeutically effective amount” means a quantity sufficient to alleviate or ameliorate one or more symptoms of a disorder, disease, or condition being treated, or to otherwise provide a desired pharmacologic and/or physiological effect. Such amelioration only requires a reduction or alteration, not necessarily elimination. The precise quantity will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, weight, etc. ) , the disease or disorder being treated, as well as the route of administration, and the pharmacokinetics and pharmacodynamics of the agent being administered.
“Microbial flora” refers to the microorganisms that normally live in the gastrointestinal tract, skin, nose, etc. In a healthy human, the internal tissues, e.g. blood, brain, muscle, etc., are normally free of microorganisms. However, the surface tissues, i.e., skin and mucous membranes, are constantly in contact with environmental organisms and become readily colonized by various microbial species. The mixture of organisms regularly found at any anatomical site is referred to as the normal flora, except by researchers in the field who prefer the term “indigenous microbiota” . Bacteria are the most numerous microbial components of the normal flora.
“Microbiota” , a term created by Jeffrey Gordon, refers to the collection of microbial species that form a microbial community. This includes the normal flora and “harmful” ones.
“Probiotic” , as used herein, utilizes the World Health Organization's 2001 definition of “live micro-organisms which, when administered in adequate amounts, confer a health benefit on the host” . Probiotics must be alive when administered, have viability and reproducibility based on in vivo testing, and during use and storage.
The term “dosage regime” refers to drug administration regarding formulation, route of administration, drug dose, dosing interval and treatment duration.
The term “pharmaceutically acceptable” or “biocompatible” refers to compositions, polymers and other materials and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase “pharmaceutically acceptable carrier” refers to pharmaceutically acceptable materials, compositions or vehicles, such as a liquid or solid filler, diluent, solvent or encapsulating material involved in carrying or transporting any subject composition, from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be acceptable” in the sense of being compatible with the other ingredients of a subject composition and not injurious to the patient.
In general, a “small molecule” is understood in the art to be an organic molecule that is less than about 2000 g/mol in size. In some forms, the small molecule is less than about 1500 g/mol or less than about 1000 g/mol. In some forms, the small molecule is less than about 800 g/mol or less than about 500 g/mol. In some forms, small molecules are non-polymeric and/or non-oligomeric. In some forms, small molecules are not proteins, peptides, or amino acids. In some forms, small molecules are not nucleic acids or nucleotides. In some forms, small molecules are not saccharides or polysaccharides.
Disclosed are materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed method and compositions. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a ligand is disclosed and discussed and a number of modifications that can be made to a number of molecules including the ligand are discussed, each and every combination and permutation of ligand and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited, each is individually and collectively contemplated. Thus, in this example, each of the combinations A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are specifically contemplated and should be considered disclosed from disclosure of A, B, and C; D, E, and F; and the example combination A-D. Likewise, any subset or combination of these is also specifically contemplated and disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E are specifically contemplated and should be considered disclosed from disclosure of A, B, and C; D, E, and F; and the example combination A-D. Further, each of the materials, compositions, components, etc. contemplated and disclosed as above can also be specifically and independently included or excluded from any group, subgroup, list, set, etc. of such materials.
These concepts apply to all aspects of this application including, but not limited to, steps in methods of making and using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific form or combination of forms of the disclosed methods, and that each such combination is specifically contemplated and should be considered disclosed.
All methods described herein can be performed in any suitable order unless otherwise indicated or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as” ) provided herein, is intended merely to better illuminate the forms and does not pose a limitation on the scope of the forms unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein.
B. Composition
Disclosed are compositions and methods for treating and/or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) and colorectal cancer.
1. Probiotic formulation
Disclosed herein are probiotic formulations, Prohep, for treating and/or preventing metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) and colorectal cancer.
The improved formulation of Prohep is listed as follow, Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium animalis subsp. lactis and Streptococcus thermophilus. And the old Prohep is composed of Lactobacillus rhamnosus GG (LGG) , viable Escherichia coli Nissle 1917 (EcN) and heat-inactivated VSL#3 (containing Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium infantis) . The detailed components comparison was illustrated in Table 3.
Table 3: Components comparison between improved and old formulation of Prohep.
The first main difference between the improved formula and old formula is that the following stains, Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium infantis are heat-inactivated in old formula, while all the stains are viable in the improved formula. Cell viability has been recognized as a crucial factor for probiotics to provide health benefits. Having multiple probiotic strains that establish themselves in the gut can enhance gut microbiota diversity and promote a more resilient and balanced gut microbiota. A recent study highlighted distinct regulatory effects between live and heat-inactivated Streptococcus thermophilus MN-ZLW-002. The live form, as opposed to the heat-inactivated form, of S. thermophilus MN-ZLW-002 can directly influence the composition of the intestinal microbiota. Therefore, multiple viable probiotic strains could imply a more profound and long-lasting effect than heat-inactivated counterpart in this case.
The second difference lays in the removal of Escherichia coli Nissle 1917 in the improved formula. Although E. coli Nissle 1917 may provide certain health benefits, the taxonomic group it belongs, Escherichia coli is increasingly involved in various intestinal and extra-intestinal infections as an opportunistic pathogen. Nonpathogenic and pathogenic strains of E. coli exhibit variations in their genetic makeup, which influence the development of specific virulence characteristics. But the genetic information responsible for these traits could be acquired through horizontal gene transfer and result in developing from commensal E. coli strains into pathogenic variants. Removal of E. coli Nissle 1917 diminishes the potential risk.
The third difference is the addition of Bifidobacterium animalis subsp. Lactis. B. animalis subsp. Lactis is a renowned probiotic strain that have been proven its beneficial health effect and safety for consumption in numerous clinical studies. Clinical studies have confirmed its positive impact on gastrointestinal health and immune function by promoting a balanced gut microbiota, enhancing bowel function, mitigating antibiotic-related side effects, and more.
The term "Prohep" as used throughout this application generally refers to an improved formulation, unless specified otherwise.
2. Anti-cancer agents
Anti-cancer agents or therapeutic agents include one or more of chemotherapeutics, targeted therapies, immunotherapies, hormonal therapies, epigenetic modulators, radiopharmaceuticals, angiogenesis inhibitors, natural products, nanoparticle-based therapies, RNA-based therapies, microbiome-modulating treatments, or a combination thereof.
Examples of anti-cancer agents that can be used in the pharmaceutical compositions containing the disclosed probiotic formulation or a separate pharmaceutical composition include, but are not limited to, temozolomide, carmustine, bevacizumab, procarbazine, lomustine, vincristine, gefitinib, erlotinib, cisplatin, carboplatin, oxaliplatin, 5-fluorouracil, gemcitabine, tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin, mithramycin, vinblastine, vindesine, vinorelbine, paclitaxel, taxol, docetaxel, etoposide, teniposide, amsacrine, topotecan, camptothecin, bortezomib, anagrelide, tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, fulvestrant, bicalutamide, flutamide, nilutamide, cyproterone, goserelin, leuprorelin, buserelin, megestrol, anastrozole, letrozole, vorozole, exemestane, finasteride, marimastat, trastuzumab, cetuximab, dasatinib, imatinib, combretastatin, thalidomide, azacitidine, azathioprine, capecitabine, chlorambucil, cyclophosphamide, cytarabine, daunorubicin, doxifluridine, epothilone, irinotecan, mechlorethamine, mercaptopurine, mitoxantrone, pemetrexed, tioguanine, valrubicin and/or lenalidomide or combinations thereof such as cyclophosphamide, methotrexate, 5-fluorouracil (CMF) ; doxorubicin, cyclophosphamide (AC) ; mustine, vincristine, procarbazine, prednisolone (MOPP) ; sdriamycin, bleomycin, vinblastine, dacarbazine (ABVD) ; cyclophosphamide, doxorubicin, vincristine, prednisolone (CHOP) ; bleomycin, etoposide, cisplatin (BEP) ; epirubicin, cisplatin, 5-fluorouracil (ECF) ; epirubicin, cisplatin, capecitabine (ECX) ; and methotrexate, vincristine, doxorubicin, cisplatin (MVAC) ; and combinations thereof. Additional anti-cancer agents are described in U.S. Patent No. 10,393,736, the disclosure of which is incorporated herein by reference in its entirety.
In some forms, the anti-cancer agent is Sorafenib. Sorafenib (brand name: Nexavar) is an oral, multi-kinase inhibitor used to treat certain cancers, including hepatocellular carcinoma (liver cancer) , renal cell carcinoma (kidney cancer) , and radioactive iodine-resistant differentiated thyroid cancer. It works by blocking specific enzymes involved in tumor cell growth and blood vessel formation, targeting pathways like VEGFR, PDGFR, and RAF kinases. By inhibiting these pathways, sorafenib slows tumor progression and reduces blood supply to tumors.
Sorafenib is a multi-kinase inhibitor that is presently utilized as a first-line therapy for advanced HCC. Nevertheless, the limited response rate, rapid development of drug resistance, and severe and extensive adverse events associated with Sorafenib are major challenges to its clinical use. Adverse events have been associated with gut dysbiosis, which results in a less protective intestinal environment and increases the incident of diarrhea while probiotics supplementation could maintain a favorable gut microbiota against Sorafenib-induced disturbances. In clinical practice, the applied dose for HCC therapy is 800 mg/day. Previously, a low dose of Sorafenib (30 mg/kg) , which is equivalent to a human dose of 135 mg/day for a 60-kg person, was supplemented in mouse and monkey models at the initial stage of MASH-HCC. Although the low dose of Sorafenib was found to effectively ameliorate major MASH hallmarks, including hepatic steatosis, inflammation, and fibrosis. However, mice treated with low-dose Sorafenib experienced slight degrees of unrecovered hair, watery stool, and diarrhea. Additionally, declining systemic drug levels were observed in the mice, indicating the onset of Sorafenib resistance.
In some forms dosage of sorafenib can range from 30 mg/kg –800 mg/kg body weight, 25 mg/kg –700 mg/kg body weight, 20 mg/kg –600 mg/kg body weight, 15 mg/kg –500 mg/kg body weight, 10 mg/kg –400 mg/kg body weight, 5 mg/kg –300 mg/kg body weight, 1 mg/kg –200 mg/kg body weight, preferably 30 mg/kg body weight. In some forms, the dosage of sorafenib is less than 30 mg/kg.
Therapeutic agents for use in the disclosed methods for treatment of the disclosed subjects are provided. The therapeutic agents are typically administered to a subject in an effective amount to treat the disease or disorder of the subject. The therapeutic agent can be in a pharmaceutical composition.
The therapeutic agent is most typically a compound that reduces the biological activity of a target molecule. Thus, compounds for decreasing the bioactivity of target molecules, and formulations formed therewith are provided. In some forms, the compound is an inhibitory polypeptide such as, but not limited to, an antibody; a small molecule or peptidomimetic, or an inhibitory nucleic acid that targets genomic or expressed nucleic acids (e.g., mRNA) encoding the target molecule, or a vector that encodes an inhibitory nucleic acid. The compound can reduce the expression or bioavailability of the target molecule. The inhibition can be competitive, non-competitive, uncompetitive, or product inhibition. Thus, an inhibitor can directly inhibit the target molecule, an inhibitor can inhibit another factor in a pathway that leads to induction, persistence, or amplification of the target molecule’s expression, or a combination thereof. Thus, the therapeutic agents can be and are also referred to herein as inhibitors.
In some forms, the therapeutic agent is a protein binder that specifically binds to the target molecule, or a ligand or receptor thereof important for activity of the target molecule. In some forms, the protein binder is an antibody. Antibodies include not only intact antibodies, but also antibody fragments and antigen-binding components thereof, and fusion proteins including antigen binding fragments that are capable of immuno-specifically binding to the target molecule (or its counterpart ligand or receipt) . The antibodies can be a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a recombinant antibody, an antigen-binding antibody fragment, a single chain antibody, a monomeric antibody, a diabody, a triabody, a tetrabody, a Fab fragment, an IgD antibody, an IgE antibody, an IgM antibody, an IgG1 antibody, an IgG2 antibody, an IgG3 antibody, and an IgG4 antibody, or a fragment thereof, and fusion proteins formed therefrom. The antibodies and antigen binding fragments can be monospecific, bispecific, trispecific or multispecific.
The inhibitor can be a functional nucleic acid. Functional nucleic acids are nucleic acid molecules that have a specific function, such as binding a target molecule or catalyzing a specific reaction. As discussed in more detail below, functional nucleic acid molecules can be divided into the following non-limiting categories: antisense molecules, siRNA, miRNA, aptamers, ribozymes, triplex forming molecules, RNAi, and external guide sequences. The functional nucleic acid molecules can act as effectors, inhibitors, modulators, and stimulators of a specific activity possessed by a target molecule, or the functional nucleic acid molecules can possess a de novo activity independent of any other molecules.
Functional nucleic acid molecules can interact with any macromolecule, such as DNA, RNA, polypeptides, or carbohydrate chains. Thus, functional nucleic acids can interact with the mRNA or the genomic DNA of a target polypeptide or they can interact with the polypeptide itself. Often functional nucleic acids are designed to interact with other nucleic acids based on sequence homology between the target molecule and the functional nucleic acid molecule. In other situations, the specific recognition between the functional nucleic acid molecule and the target molecule is not based on sequence homology between the functional nucleic acid molecule and the target molecule, but rather is based on the formation of tertiary structure that allows specific recognition to take place.
Therefore, the compositions can include one or more functional nucleic acids designed to reduce expression of the target molecule’s gene, or a gene product thereof. For example, the functional nucleic acid or polypeptide can be designed to target and reduce or inhibit expression or translation of target molecule’s mRNA; or to reduce or inhibit expression, reduce activity, or increase degradation of target molecule protein. In some forms, the composition includes a vector suitable for in vivo expression of the functional nucleic acid.
Examples of functional nucleic acids include, but are not limited to, antisense oligonucleotides, siRNA, shRNA, miRNA, external guide sequences. External guide sequences (EGSs) , ribozymes, aptamers, and CRISPR/Cas technology.
C. Other formulations
The disclosed compounds can be formulated in a pharmaceutical composition. Pharmaceutical compositions can be for administration by oral route, parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection) , enteral, transdermal (either passively or using iontophoresis or electroporation) , or transmucosal (nasal, pulmonary, vaginal, rectal, or sublingual) routes of administration or using bioerodible inserts and can be formulated in dosage forms appropriate for each route of administration.
The compositions can be administered systemically.
Drugs can be formulated for immediate release, extended release, or modified release. A delayed release dosage form is one that releases a drug (or drugs) at a time other than promptly after administration. An extended release dosage form is one that allows at least a twofold reduction in dosing frequency as compared to that drug presented as a conventional dosage form (e.g. as a solution or prompt drug-releasing, conventional solid dosage form) . A modified release dosage form is one for which the drug release characteristics of time course and/or location are chosen to accomplish therapeutic or convenience objectives not offered by conventional dosage forms such as solutions, ointments, or promptly dissolving dosage forms. Delayed release and extended release dosage forms and their combinations are types of modified release dosage forms.
Formulations are typically prepared using a pharmaceutically acceptable “carrier” composed of materials that are considered safe and effective and may be administered to an individual without causing undesirable biological side effects or unwanted interactions. The “carrier” is all components present in the pharmaceutical formulation other than the active ingredient or ingredients. The term “carrier” includes, but is not limited to, diluents, binders, lubricants, disintegrators, fillers, and coating compositions.
“Carrier” also includes all components of the coating composition which may include plasticizers, pigments, colorants, stabilizing agents, and glidants. The delayed release dosage formulations may be prepared as described in references such as “Pharmaceutical dosage form tablets” , eds. Liberman et al. (New York, Marcel Dekker, Inc., 1989) , “Remington –The science and practice of pharmacy” , 20th ed., Lippincott Williams &Wilkins, Baltimore, MD, 2000, and “Pharmaceutical dosage forms and drug delivery systems” , 6th Edition, Ansel et.al., (Media, PA: Williams and Wilkins, 1995) which provides information on carriers, materials, equipment and process for preparing tablets and capsules and delayed release dosage forms of tablets, capsules, and granules.
The compound can be administered to a subject with or without the aid of a delivery vehicle. Appropriate delivery vehicles for the compounds are known in the art and can be selected to suit the particular active agent. For example, in some forms, the active agent (s) is incorporated into or encapsulated by, or bound to, a nanoparticle, microparticle, micelle, synthetic lipoprotein particle, or carbon nanotube. For example, the compositions can be incorporated into a vehicle such as polymeric particles which provide controlled release of the active agent (s) . In some forms, release of the drug (s) is controlled by diffusion of the active agent (s) out of the particles and/or degradation of the polymeric particles by hydrolysis and/or enzymatic degradation.
Suitable polymers include ethylcellulose and other natural or synthetic cellulose derivatives. Polymers which are slowly soluble and form a gel in an aqueous environment, such as hydroxypropyl methylcellulose or polyethylene oxide, may also be suitable as materials for drug containing particles or particles. Other polymers include, but are not limited to, polyanhydrides, poly (ester anhydrides) , polyhydroxy acids, such as polylactide (PLA) , polyglycolide (PGA) , poly (lactide-co-glycolide) (PLGA) , poly-3-hydroxybut rate (PHB) and copolymers thereof, poly-4-hydroxybutyrate (P4HB) and copolymers thereof, polycaprolactone and copolymers thereof, and combinations thereof. In some forms, both agents are incorporated into the same particles and are formulated for release at different times and/or over different time periods. For example, in some forms, one of the agents is released entirely from the particles before release of the second agent begins. In other forms, release of the first agent begins followed by release of the second agent before the all of the first agent is released. In still other forms, both agents are released at the same time over the same period of time or over different periods of time.
Suitable pharmaceutically acceptable carriers and excipients are generally recognized as safe (GRAS) , and may be administered to an individual without causing undesirable biological side effects or unwanted interactions.
Representative carriers and excipients include solvents (including buffers) , diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agents, viscosity modifiers, tonicity agents, and stabilizing agents, and a combination thereof.
Excipients can be added to a liquid or solid pharmaceutical composition (for in vivo or in vitro applications) to assist in sterility, stability (e.g. shelf-life) , integration, and to adjust and/or maintain pH or isotonicity of the inhibitors in the pharmaceutical composition, such as diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agents, viscosity modifiers, tonicity agents, and stabilizing agents, and a combination thereof.
1. Oral formulation
Suitable oral dosage forms include tablets, capsules, solutions, suspensions, syrups, and lozenges. Tablets can be made using compression or molding techniques well known in the art. Gelatin or non-gelatin capsules can prepared as hard or soft capsule shells, which can encapsulate liquid, solid, and semi-solid fill materials, using techniques well known in the art.
Examples of suitable coating materials include, but are not limited to, cellulose polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate and hydroxypropyl methylcellulose acetate succinate; polyvinyl acetate phthalate, acrylic acid polymers and copolymers, and methacrylic resins that are commercially available under the trade name (Roth Pharma, Westerstadt, Germany) , Zein, shellac, and polysaccharides.
Additionally, the coating material may contain conventional carriers such as plasticizers, pigments, colorants, glidants, stabilization agents, pore formers and surfactants.
Optional pharmaceutically acceptable excipients present in the drug-containing tablets, beads, granules or particles include, but are not limited to, diluents, binders, lubricants, disintegrants, colorants, stabilizers, and surfactants. Diluents, also termed "fillers, " are typically necessary to increase the bulk of a solid dosage form so that a practical size is provided for compression of tablets or formation of beads and granules. Suitable diluents include, but are not limited to, , dicalcium phosphate dihydrate, calcium sulfate, lactose, sucrose, mannitol, sorbitol, cellulose, microcrystalline cellulose, kaolin, sodium chloride, dry starch, hydrolyzed starches, pregelatinized starch, silicone dioxide, titanium oxide, magnesium aluminum silicate and powder sugar.
Binders are used to impart cohesive qualities to a solid dosage formulation and thus ensure that a tablet or bead or granule remains intact after the formation of the dosage forms. Suitable binder materials include, but are not limited to, starch, pregelatinized starch, gelatin, sugars (including sucrose, glucose, dextrose, lactose and sorbitol) , polyethylene glycol, waxes, natural and synthetic gums such as acacia, tragacanth, sodium alginate, cellulose, including hydorxypropylmethylcellulose, hydroxypropylcellulose, ethylcellulose, and veegum, and synthetic polymers such as acrylic acid and methacrylic acid copolymers, methacrylic acid copolymers, methyl methacrylate copolymers, aminoalkyl methacrylate copolymers, polyacrylic acid/polymethacrylic acid and polyvinylpyrrolidone.
Lubricants are used to facilitate tablet manufacture. Examples of suitable lubricants include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, glycerol behenate, polyethylene glycol, talc, and mineral oil.
Disintegrants are used to facilitate dosage form disintegration or "breakup" after administration, and generally include, but are not limited to, starch, sodium starch glycolate, sodium carboxymethyl starch, sodium carboxymethylcellulose, hydroxypropyl cellulose, pregelatinized starch, clays, cellulose, alginine, gums or cross-linked polymers, such as cross-linked PVP (Polyplasdone XL from GAF Chemical Corp) .
Stabilizers are used to inhibit or retard drug decomposition reactions which include, by way of example, oxidative reactions.
Surfactants may be anionic, cationic, amphoteric or nonionic surface-active agents. Suitable anionic surfactants include, but are not limited to, those containing carboxylate, sulfonate and sulfate ions. Examples of anionic surfactants include sodium, potassium, ammonium of long chain alkyl sulfonates and alkyl aryl sulfonates such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium dodecylbenzene sulfonate; dialkyl sodium sulfosuccinates, such as sodium bis- (2-ethylthioxyl) -sulfosuccinate; and alkyl sulfates such as sodium lauryl sulfate. Cationic surfactants include, but are not limited to, quaternary ammonium compounds such as benzalkonium chloride, benzethonium chloride, cetrimonium bromide, stearyl dimethylbenzyl ammonium chloride, polyoxyethylene and coconut amine. Examples of nonionic surfactants include ethylene glycol monostearate, propylene glycol myristate, glyceryl monostearate, glyceryl stearate, polyglyceryl-4-oleate, sorbitan acylate, sucrose acylate, PEG-150 laurate, PEG-400 monolaurate, polyoxyethylene monolaurate, polysorbates, polyoxyethylene octylphenylether, PEG-1000 cetyl ether, polyoxyethylene tridecyl ether, polypropylene glycol butyl ether, 401, stearoyl monoisopropanolamide, and polyoxyethylene hydrogenated tallow amide. Examples of amphoteric surfactants include sodium N-dodecyl-. beta. -alanine, sodium N-lauryl-. beta. -iminodipropionate, myristoamphoacetate, lauryl betaine and lauryl sulfobetaine.
If desired, the tablets, beads granules or particles may also contain minor amount of nontoxic auxiliary substances such as wetting or emulsifying agents, dyes, pH buffering agents, and preservatives.
2. Extended release dosage forms
The extended release formulations are generally prepared as diffusion or osmotic systems, for example, as described in “Remington –The science and practice of pharmacy” (20th ed., Lippincott Williams &Wilkins, Baltimore, MD, 2000) . A diffusion system typically consists of two types of devices, reservoir and matrix, and is well known and described in the art. The matrix devices are generally prepared by compressing the drug with a slowly dissolving polymer carrier into a tablet form. The three major types of materials used in the preparation of matrix devices are insoluble plastics, hydrophilic polymers, and fatty compounds. Plastic matrices include, but not limited to, methyl acrylate-methyl methacrylate, polyvinyl chloride, and polyethylene. Hydrophilic polymers include, but are not limited to, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and carbopol 934, polyethylene oxides. Fatty compounds include, but are not limited to, various waxes such as carnauba wax and glyceryl tristearate.
Alternatively, extended release formulations can be prepared using osmotic systems or by applying a semi-permeable coating to the dosage form. In the latter case, the desired drug release profile can be achieved by combining low permeable and high permeable coating materials in suitable proportion.
The devices with different drug release mechanisms described above could be combined in a final dosage form having single or multiple units. Examples of multiple units include multilayer tablets, capsules containing tablets, beads, granules, etc.
An immediate release portion can be added to the extended release system by means of either applying an immediate release layer on top of the extended release core using coating or compression process or in a multiple unit system such as a capsule containing extended and immediate release beads.
Extended release tablets containing hydrophilic polymers are prepared by techniques commonly known in the art such as direct compression, wet granulation, or dry granulation processes. Their formulations usually incorporate polymers, diluents, binders, and lubricants as well as the active pharmaceutical ingredient. The usual diluents include inert powdered substances such as any of many different kinds of starch, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives are also useful. Typical tablet binders include substances such as starch, gelatin and sugars such as lactose, fructose, and glucose. Natural and synthetic gums, including acacia, alginates, methylcellulose, and polyvinylpyrrolidine can also be used. Polyethylene glycol, hydrophilic polymers, ethylcellulose and waxes can also serve as binders. A lubricant is necessary in a tablet formulation to prevent the tablet and punches from sticking in the die. The lubricant is chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils.
Extended release tablets containing wax materials are generally prepared using methods known in the art such as a direct blend method, a congealing method, and an aqueous dispersion method. In a congealing method, the drug is mixed with a wax material and either spray-congealed or congealed and screened and processed.
3. Delayed release dosage forms
Delayed release formulations are created by coating a solid dosage form with a film of a polymer which is insoluble in the acid environment of the stomach, and soluble in the neutral environment of small intestines.
The delayed release dosage units can be prepared, for example, by coating a drug or a drug-containing composition with a selected coating material. The drug-containing composition may be, e.g., a tablet for incorporation into a capsule, a tablet for use as an inner core in a "coated core" dosage form, or a plurality of drug-containing beads, particles or granules, for incorporation into either a tablet or capsule. Preferred coating materials include bioerodible, gradually hydrolyzable, gradually water-soluble, and/or enzymatically degradable polymers, and may be conventional "enteric" polymers. Enteric polymers, as will be appreciated by those skilled in the art, become soluble in the higher pH environment of the lower gastrointestinal tract or slowly erode as the dosage form passes through the gastrointestinal tract, while enzymatically degradable polymers are degraded by bacterial enzymes present in the lower gastrointestinal tract, particularly in the colon. Suitable coating materials for effecting delayed release include, but are not limited to, cellulosic polymers such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxymethyl cellulose, hydroxypropyl methyl cellulose, hydroxypropyl methyl cellulose acetate succinate, hydroxypropylmethyl cellulose phthalate, methylcellulose, ethyl cellulose, cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate and carboxymethylcellulose sodium; acrylic acid polymers and copolymers, preferably formed from acrylic acid, methacrylic acid, methyl acrylate, ethyl acrylate, methyl methacrylate and/or ethyl methacrylate, and other methacrylic resins that are commercially available under the tradename(Rohm Pharma; Westerstadt, Germany) , includingL30D-55 and L100-55 (soluble at pH 5.5 and above) , L-100 (soluble at pH 6.0 and above) , S(soluble at pH 7.0 and above, as a result of a higher degree of esterification) , andNE, RL and RS (water-insoluble polymers having different degrees of permeability and expandability) ; vinyl polymers and copolymers such as polyvinyl pyrrolidone, vinyl acetate, vinylacetate phthalate, vinylacetate crotonic acid copolymer, and ethylene-vinyl acetate copolymer; enzymatically degradable polymers such as azo polymers, pectin, chitosan, amylose and guar gum; zein and shellac. Combinations of different coating materials may also be used. Multi-layer coatings using different polymers may also be applied.
The preferred coating weights for particular coating materials may be readily determined by those skilled in the art by evaluating individual release profiles for tablets, beads and granules prepared with different quantities of various coating materials. It is the combination of materials, method and form of application that produce the desired release characteristics, which one can determine only from the clinical studies.
The coating composition may include conventional additives, such as plasticizers, pigments, colorants, stabilizing agents, glidants, etc. A plasticizer is normally present to reduce the fragility of the coating, and will generally represent about 10 wt. %to 50 wt. %relative to the dry weight of the polymer. Examples of typical plasticizers include polyethylene glycol, propylene glycol, triacetin, dimethyl phthalate, diethyl phthalate, dibutyl phthalate, dibutyl sebacate, triethyl citrate, tributyl citrate, triethyl acetyl citrate, castor oil and acetylated monoglycerides. A stabilizing agent is preferably used to stabilize particles in the dispersion. Typical stabilizing agents are nonionic emulsifiers such as sorbitan esters, polysorbates and polyvinylpyrrolidone. Glidants are recommended to reduce sticking effects during film formation and drying, and will generally represent approximately 25 wt. %to 100 wt. %of the polymer weight in the coating solution. One effective glidant is talc. Other glidants such as magnesium stearate and glycerol monostearates may also be used. Pigments such as titanium dioxide may also be used. Small quantities of an anti-foaming agent, such as a silicone (e.g., simethicone) , may also be added to the coating composition.
As will be appreciated by those skilled in the art and as described in the pertinent texts and literature, a number of methods are available for preparing drug-containing tablets, beads, granules or particles that provide a variety of drug release profiles. Such methods include, but are not limited to, the following: coating a drug or drug-containing composition with an appropriate coating material, typically although not necessarily incorporating a polymeric material, increasing drug particle size, placing the drug within a matrix, and forming complexes of the drug with a suitable complexing agent.
The delayed release dosage units may be coated with the delayed release polymer coating using conventional techniques, e.g., using a conventional coating pan, an airless spray technique, fluidized bed coating equipment (with or without a Wurster insert) . For detailed information concerning materials, equipment and processes for preparing tablets and delayed release dosage forms, see Pharmaceutical Dosage Forms: Tablets, eds. Lieberman et al. (New York: Marcel Dekker, Inc., 1989) , and Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 6. sup. th Ed. (Media, PA: Williams &Wilkins, 1995) .
A method for preparing extended release tablets is by compressing a drug-containing blend, e.g., blend of granules, prepared using a direct blend, wet-granulation, or dry-granulation process. Extended release tablets may also be molded rather than compressed, starting with a moist material containing a suitable water-soluble lubricant. However, tablets are preferably manufactured using compression rather than molding. A preferred method for forming extended release drug-containing blend is to mix drug particles directly with one or more excipients such as diluents (or fillers) , binders, disintegrants, lubricants, glidants, and colorants. As an alternative to direct blending, a drug-containing blend may be prepared by using wet-granulation or dry-granulation processes. Beads containing the active agent may also be prepared by any one of a number of conventional techniques, typically starting from a fluid dispersion. For example, a typical method for preparing drug-containing beads involves dispersing or dissolving the active agent in a coating suspension or solution containing pharmaceutical excipients such as polyvinylpyrrolidone, methylcellulose, talc, metallic stearates, silicone dioxide, plasticizers or the like. The admixture is used to coat a bead core such as a sugar sphere (or so-called "non-pareil" ) having a size of approximately 60 to 20 mesh.
An alternative procedure for preparing drug beads is by blending drug with one or more pharmaceutically acceptable excipients, such as microcrystalline cellulose, lactose, cellulose, polyvinyl pyrrolidone, talc, magnesium stearate, a disintegrant, etc., extruding the blend, spheronizing the extrudate, drying and optionally coating to form the immediate release beads.
4. Mucosal formulation
The disclosed composition can be formulated for mucosal administration.
In some forms, the disclosed composition is formulated for direct application on the mucous membrane generally contain a dermatologically acceptable carrier that is suitable for application to the mucous membrane, has good aesthetic properties, is compatible with the active agents and any other components, and will not cause any untoward safety or toxicity concerns.
The carrier can be in a wide variety of forms. For example, emulsion carriers, including, but not limited to, oil-in-water, water-in-oil, water-in-oil-in-water, and oil-in-water-in-silicone emulsions, are useful herein. These emulsions can cover a broad range of viscosities, e.g., from about 100 cps to about 200,000 cps. These emulsions can also be delivered in the form of sprays using either mechanical pump containers or pressurized aerosol containers using conventional propellants. These carriers can also be delivered in the form of a mousse or a transdermal patch. Other suitable topical carriers include anhydrous liquid solvents such as oils, alcohols, and silicones (e.g., mineral oil, ethanol isopropanol, dimethicone, cyclomethicone, and the like) ; aqueous-based single phase liquid solvents (e.g., hydro-alcoholic solvent systems, such as a mixture of ethanol and/or isopropanol and water) ; and thickened versions of these anhydrous and aqueous-based single phase solvents (e.g. where the viscosity of the solvent has been increased to form a solid or semi-solid by the addition of appropriate gums, resins, waxes, polymers, salts, and the like) . Examples of topical carrier systems useful in the present formulations are described in the following four references all of which are incorporated herein by reference in their entirety: “Sun Products Formulary” Cosmetics &Toiletries, vol. 105, pp. 122-139 (December 1990) ; “Sun Products Formulary, ” Cosmetics &Toiletries, vol. 102, pp. 117-136 (March 1987) ; U.S. Pat. No. 5,605,894 to Blank et al., and U.S. Pat. No. 5,681,852 to Bissett.
Formulations for direct application on the mucous membrane may be formulated to be immediate and/or modified release. Modified release formulations include delayed, sustained, pulsed, controlled, targeted and programmed release formulations. Thus, the disclosed composition may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active agents.
5. Parenteral Formulations
The disclosed composition can be formulated for Parenteral administration. The disclosed composition can be formulated in a form suitable for administration directly into the blood stream, into muscle, or into an internal organ. Suitable routes for such parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, epidural, intracerebroventricular, intraurethral, intrasternal, intracranial, intramuscular, and subcutaneous delivery. Suitable means for parenteral administration include needle (including microneedle) injectors, needle free injectors, and infusion techniques.
For example, the disclosed composition can be formulated in a form suitable for intramuscular administration, intravenous administration, intraperitoneal administration, or subcutaneous administration, or a combination thereof.
In some forms, the disclosed composition described herein can be in aqueous solutions which can contain excipients such as salts, carbohydrates and buffering agents (e.g., from about pH 6.5 to about pH 8.0, from about pH 6.5 to about pH 7.4, from about pH 6.5 to about pH 7.0, from about pH 7.0 to pH 8.0, or from about pH 7.0 to about pH 7.4) , but, for some applications, they may be more suitably formulated as a sterile aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen free water.
In some forms, the disclosed composition may be a solution, a suspension, or an emulsion. In some forms, the disclosed composition can include one or more physiologically compatible buffers, such as a phosphate buffers. One skilled in the art can readily determine a suitable saline content and pH for an aqueous carrier for administration (e.g., from about pH 6.5 to about pH 8.0, from about pH 6.5 to about pH 7.4, from about pH 6.5 to about pH 7.0, from about pH 7.0 to pH 8.0, or from about pH 7.0 to about pH 7.4) .
In some forms, the disclosed composition for parenteral administration may include one or more suspending agents, such as cellulose derivatives, sodium alginate, polyvinylpyrrolidone, gum tragacanth, or lecithin. The liquid compositions may also include one or more preservatives, such as ethyl or n-propyl p-hydroxybenzoate.
In some forms, the disclosed composition may contain one or more solvents that are low toxicity organic (i.e., nonaqueous) class 3 residual solvents, such as ethanol, acetone, ethyl acetate, tetrahydofuran, ethyl ether, and propanol, and a combination thereof. Solvents such as freon, alcohol, glycol, polyglycol, or fatty acid, can also be included in the liquid composition.
In some forms, the disclosed composition may also contain minor amounts of polymers, surfactants, or other pharmaceutically acceptable excipients known to those in the art. In some forms, the disclosed composition is formulated typically under sterile conditions, for example, by lyophilisation, which can be accomplished using standard pharmaceutical techniques known to those skilled in the art.
In some forms, the disclosed composition may be formulated to provide immediate and/or modified release. Modified release formulations include delayed, sustained, pulsed, controlled, targeted and programmed release formulations.
D. Methods
Methods of treating a subject in need thereof are provided.
1. Diseases to be treated
Methods of treating diseases and/or disorders in a subject in need thereof are provided. The subject to be treated can have a disease, disorder, or condition such as but not limited to, cancer, an immune system disorder such autoimmune disease, an inflammatory disease, a neuronal disorder, HIV/AIDS, diabetes, a cardiovascular disease, an infectious disease, or combinations thereof.
i. Cancer
In some forms, the methods treat or prevent cancer. In some forms, the methods treat or prevent metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) . In some forms, the methods treat or prevent colorectal cancer (CRC) .
In some forms, the methods treat or prevent cancer or other proliferative disease or disorder in a subject identified as having, or at risk of having cancer or other proliferative disease or disorder. Cancer is a disease of genetic instability, allowing a cancer cell to acquire the hallmarks proposed by Hanahan and Weinberg, including (i) self-sufficiency in growth signals; (ii) insensitivity to anti-growth signals; (iii) evading apoptosis; (iv) sustained angiogenesis; (v) tissue invasion and metastasis; (vi) limitless replicative potential; (vii) reprogramming of energy metabolism; and (viii) evading immune destruction (Cell., 144: 646–674, (2011) ) .
Tumors, which can be treated in accordance with the disclosed methods, are classified according to the embryonic origin of the tissue from which the tumor is derived. Carcinomas are tumors arising from endodermal or ectodermal tissues such as skin or the epithelial lining of internal organs and glands. Sarcomas, which arise less frequently, are derived from mesodermal connective tissues such as bone, fat, and cartilage. The leukemias and lymphomas are malignant tumors of hematopoietic cells of the bone marrow. Leukemias proliferate as single cells, whereas lymphomas tend to grow as tumor masses. Malignant tumors may show up at numerous organs or tissues of the body to establish a cancer.
Table 5: The disclosed compositions and methods can be used in the treatment of one or more cancers.


The disclosed compositions and methods of treatment thereof are generally suited for treatment of carcinomas, sarcomas, lymphomas and leukemias. The described compositions and methods are useful for treating, or alleviating subjects having benign or malignant tumors by delaying or inhibiting the growth/proliferation or viability of tumor cells in a subject, reducing the number, growth or size of tumors, inhibiting or reducing metastasis of the tumor, and/or inhibiting or reducing symptoms associated with tumor development or growth.
The types of cancer that can be treated with the provided compositions and methods include, but are not limited to, cancers such as vascular cancer such as multiple myeloma, adenocarcinomas and sarcomas, of bone, bladder, brain, breast, cervical, colorectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, and uterine. The experiments below support the conclusion that the disclosure approach is effective for treating solid tumors. Thus, in some forms, the target cancer is a solid tumor. In some forms, the compositions are used to treat multiple cancer types concurrently. The compositions can also be used to treat metastases or tumors at multiple locations.
Exemplary tumor cells include, but are not limited to, tumor cells of cancers, including leukemias including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome, chronic leukemias such as, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as, but not limited to, Hodgkin’s disease, non-Hodgkin’s disease; multiple myelomas such as, but not limited to, smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma and extramedullary plasmacytoma; macroglobulinemia; monoclonal gammopathy of undetermined significance; benign monoclonal gammopathy; heavy chain disease; bone and connective tissue sarcomas such as, but not limited to, bone sarcoma, osteosarcoma, chondrosarcoma, Ewing’s sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma) , fibrosarcoma, Kaposi’s sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, neurilemmoma, rhabdomyosarcoma, synovial sarcoma; brain tumors including, but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, primary brain lymphoma; breast cancer including, but not limited to, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, Paget’s disease, and inflammatory breast cancer; adrenal cancer, including, but not limited to, pheochromocytom and adrenocortical carcinoma; thyroid cancer such as but not limited to papillary or follicular thyroid cancer, medullary thyroid cancer and anaplastic thyroid cancer; pancreatic cancer, including, but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; pituitary cancers including, but not limited to, Cushing’s disease, prolactin-secreting tumor, acromegaly, and diabetes insipius; eye cancers including, but not limited to, ocular melanoma such as iris melanoma, choroidal melanoma, and ciliary body melanoma, and retinoblastoma; vaginal cancers, including, but not limited to, squamous cell carcinoma, adenocarcinoma, and melanoma; vulvar cancer, including, but not limited to, squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget’s disease; cervical cancers including, but not limited to, squamous cell carcinoma, and adenocarcinoma; uterine cancers including, but not limited to, endometrial carcinoma and uterine sarcoma; ovarian cancers including, but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor; esophageal cancers including, but not limited to, squamous cancer, adenocarcinoma, adenoid cyctic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma; stomach cancers including, but not limited to, adenocarcinoma, fungating (polypoid) , ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; colon cancers; rectal cancers; liver cancers including, but not limited to, hepatocellular carcinoma and hepatoblastoma, gallbladder cancers including, but not limited to, adenocarcinoma; cholangiocarcinomas including, but not limited to, papillary, nodular, and diffuse; lung cancers including, but not limited to, non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma) , adenocarcinoma, large-cell carcinoma and small-cell lung cancer; testicular cancers including, but not limited to, germinal tumor, seminoma, anaplastic, classic (typical) , spermatocytic, nonseminoma, embryonal carcinoma, teratoma carcinoma, choriocarcinoma (yolk-sac tumor) , prostate cancers including, but not limited to, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; penal cancers; oral cancers including, but not limited to, squamous cell carcinoma; basal cancers; salivary gland cancers including, but not limited to, adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; pharynx cancers including, but not limited to, squamous cell cancer, and verrucous; skin cancers including, but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, acral lentiginous melanoma; kidney cancers including, but not limited to, renal cell cancer, adenocarcinoma, hypernephroma, fibrosarcoma, transitional cell cancer (renal pelvis and/or uterer) ; Wilms’ tumor; bladder cancers including, but not limited to, transitional cell carcinoma, squamous cell cancer, adenocarcinoma, and carcinosarcoma. For a review of such disorders, see Fishman et al., 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al., 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America) .
E. Combination therapy
The disclosed composition and methods for treating metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) or colorectal cancer (CRC) can be used in combination with other therapeutic agents or treatment modalities.
The disclosed composition and methods for treating metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) or colorectal cancer (CRC) can be used in combination with other therapeutic agents or treatment modalities such as chemotherapy, stem-cell transplantation, or immunotherapy.
As used herein, “combination” or “combined” refer to either concomitant, simultaneous, or sequential administration of the therapeutics.
In some forms, the pharmaceutical compositions and other therapeutic agents are administered separately through the same route of administration. In other forms, the pharmaceutical compositions and other therapeutic agents are administered separately through different routes of administration. The combinations can be administered either concomitantly (e.g., as an admixture) , separately but simultaneously (e.g., via separate intravenous lines into the same subject; one agent is given orally while the other agent is given by infusion or injection, etc., ) , or sequentially (e.g., one agent is given first followed by the second) .
The compositions and methods described herein may be used as a first therapy, second therapy, third therapy, or combination therapy with other types of therapies known in the art, such as chemotherapy, surgery, radiation, gene therapy, immunotherapy, bone marrow transplantation, stem cell transplantation, targeted therapy, cryotherapy, ultrasound therapy, photodynamic therapy, radio-frequency ablation or the like, in an adjuvant setting or a neoadjuvant setting.
The disclosed pharmaceutical compositions and/or other therapeutic agents, procedures or modalities can be administered during periods of active disease, or during a period of remission or less active disease. The pharmaceutical compositions can be administered before the additional treatment, concurrently with the treatment, post-treatment, or during remission of the disease or disorder. When administered in combination, the disclosed pharmaceutical compositions and the additional therapeutic agents (e.g., second or third agent) , or all, can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy. In certain forms, the administered amount or dosage of the disclosed pharmaceutical composition, the additional therapeutic agent (e.g., second or third agent) , or all, is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy (e.g., required to achieve the same therapeutic effect) .
In some forms, examples of additional therapeutic agents include other conventional therapies known in the art for treating the desired disease, disorder or condition. In some forms, the therapeutic agent is one or more other targeted therapies (e.g., a targeted cancer therapy) and/or immune-checkpoint blockage agents (e.g., anti-LAG-3, anti CTLA 4, anti PD1, and/or anti PDL1 agents such as antibodies) .
F. Dosage Units and Methods of Administration
A treatment regimen can include one or multiple administrations of the compositions including an effective amount of one or more of the compounds for achieving a desired physiological change, including administering to an animal, such as a mammal, especially a human being, an effective amount of the compositions to treat the disease or symptom thereof, or to produce the physiological change.
The effective amount or therapeutically effective amount of a pharmaceutical compositions can be a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of a disease or disorder or to otherwise provide a desired pharmacologic and/or physiologic effect, for example, reducing, inhibiting, or reversing one or more of the underlying pathophysiological mechanisms underlying a disease or disorder, such as PCAD. In preferred forms, the desired physiological change could include improvement in one or more symptoms of a disease or condition treated herein, such as improvement in breathing and exercise capacity or improved sensitivity to irritants or drop in albuterol use or improved vocal cord function or reduction in cough in the subject.
In some forms, when administrating the pharmaceutical composition, the amount administered can be expressed as the amount effective to achieve a desired effect in the recipient.
The effective amount of the pharmaceutical composition will vary based on the active agent and from subject to subject, depending on the species, age, weight and general condition of the subject, the severity of the disorder being treated, and its mode of administration. Thus, it is not possible to specify an exact amount for every pharmaceutical composition. However, an appropriate amount can be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. For example, effective dosages and schedules for administering the pharmaceutical composition can be determined empirically. In some forms, the dosage ranges for the administration of the composition are those large enough to resolve mucosal hyperreactivity throughout the respiratory tract.
Preferably, the dosage is not so large as to cause adverse side effects, such as unwanted cross-reactions, anaphylactic reactions, and the like. Generally, the dosage will vary with the age, condition, and sex of the patient, route of administration, whether other drugs are included in the regimen, and the type, stage, and location of the disease to be treated. The dosage can be adjusted by the individual physician in the event of any counter-indications. It will also be appreciated that the effective dosage of the composition can increase or decrease over the course of a particular treatment. Changes in dosage can result and become apparent from the results of diagnostic assays.
Dosage can vary, and can be administered in one or more dose administrations daily, for one or several days. Guidance can be found in the literature for appropriate dosages for given classes of pharmaceutical products. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the subject or patient. Persons of ordinary skill can determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages can vary depending on the relative potency of individual pharmaceutical compositions, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models.
In cases of a solid dosage form, examples of daily dosages of the compounds described herein which can be used are an effective amount within the dosage range of about 0.001 mg to about 2 mg per kilogram of body weight, about 0.001 mg to about 5 mg per kilogram of body weight, about 0.001 mg to about 10 mg per kilogram of body weight, about 0.001 mg to about 20 mg per kilogram of body weight, about 0.001 mg to about 50 mg per kilogram of body weight, about 0.001 mg to about 100 mg per kilogram of body weight, about 0.001 mg to about 200 mg per kilogram of body weight, or about 0.001 mg to about 300 mg per kilogram of body weight.
When administered orally, examples of daily dosages are an effective amount within the dosage range of about 0.1 mg to about 10 mg, or about 0.1 mg to about 20 mg, or about 0.1 mg to about 30 mg, or about 0.1 mg to about 40 mg, or about 0.1 mg to about 50 mg, or about 0.1 mg to about 60 mg, or about 0.1 mg to about 70 mg, or about 0.1 mg to about 80 mg, or about 0.1 mg to about 90 mg, or about 0.1 mg to about 100 mg, or about 0.1 mg to about 200 mg, or about 0.1 mg to about 300 mg, or about 0.1 mg to about 400 mg, or about 0.1 mg to about 500 mg, or about 0.1 mg to about 600 mg, or about 0.1 mg to about 700 mg, or about 0.1 mg to about 800 mg, or about 0.1 mg to about 900 mg, or about 0.1 mg to about 1 g, or about 20 mg to 300 mg, or about 20 mg to 500 mg, or about 20 mg to 700 mg, or about 20 mg to 1000 mg, or about 50 mg to 1500 mg, or about 50 mg to 2000 mg.
Exemplary fixed daily doses include about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 12 mg, about 15 mg, about 18 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 700 mg, about 800 mg, about 900 mg, about 1000 mg, about 1200 mg, about 1500 mg, or about 2000 mg, independently of body weight. However, it is understood that pediatric patients may require smaller dosages, and depending on the severity of the disease and condition of the patient, dosages may vary.
When formulated as a liquid, the concentration of the compounds described herein may be about 0.01 mg/ml to about 0.1 mg/ml or about 0.1 mg/ml to about 1 mg/ml, but can also be about 1 mg/ml to about 10 mg/ml or about 10 mg/ml to about 100 mg/ml. The liquid formulation could be a solution or a suspension. When formulated as a solid, for example as a tablet or as a powder for inhalation, the concentration, expressed as the weight of a compound divided by total weight, will typically be about 0.01%to about 0.1%, about 0.1%to about 1%, about 1%to about 10%, about 10%to about 20%, about 20%to about 40%, about 40%to about 60%, about 60%to about 80%, or about 80%to about 100%.
In some forms, administration of the composition will be given as a long-term treatment regimen whereby pharmacokinetic steady state conditions will be reached.
Injections and infusion of the disclosed compositions can be repeated as often and as many times as the patient can tolerate until the desired response is achieved. The optimal dosage and treatment regime for a particular patient can be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly. In some forms, the unit dosage is in a unit dosage form for intravenous injection. In some forms, the unit dosage is in a unit dosage form for oral administration. In some forms, the unit dosage is in a unit dosage form for inhalation. In some forms, the unit dosage is in a unit dosage form for subcutaneous injection.
Treatment can be continued for an amount of time sufficient to achieve one or more desired therapeutic goals. The timing of the administration of the composition will also depend on the formulation and/or route of administration used. The compound may be administered once daily, but may also be administered two, three or four times daily, or every other day, or once or twice per week. For example, the subject can be administered one or more treatments 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, days, weeks, or months apart.
Treatment can be continued for a desired period of time, and the progression of treatment can be. In some forms, administration is carried out every day of treatment, or every week, or every fraction of a week. In some forms, treatment regimens are carried out over the course of up to two, three, four or five days, weeks, or months, or for up to 6 months, or for more than 6 months, for example, up to one year, two years, three years, or up to five years.
The efficacy of administration of a particular dose of the pharmaceutical compositions can be determined by evaluating the aspects of the medical history, signs, symptoms, and objective laboratory tests that are known to be useful in evaluating the status of a subject in need for the treatment of a disease or condition discussed herein. These signs, symptoms, and objective laboratory tests will vary, depending upon the particular disease or condition being treated or prevented, as will be known to any clinician who treats such patients or a researcher conducting experimentation in this field.
It is to be understood that the disclosed method and compositions are not limited to specific synthetic methods, specific analytical techniques, or to particular reagents unless otherwise specified, and, as such, can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
Examples
Example 1: Probiotics mixture, prohep, is an adjuvant for low-dose sorafenib in metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma treatment through modulating gut microbiota.
Materials and methods
Probiotic Mixture Composition
Prohep is a proprietary probiotic mixture developed by the inventors of this application (IP01473) . The modified formula was composed of Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium lactis and Streptococcus thermophilus in lyophilized powder, produced under GMP (Fukopharma, Finland) as previously reported [Zhang, F., et al., Probiotic Mixture Ameliorates a Diet-Induced MASLD/MASH Murine Model through the Regulation of Hepatic Lipid Metabolism and the Gut Microbiome. J Agric Food Chem, 2024.72 (15) : p. 8536-8549] .
MASLD-HCC mouse model
Two weeks old male C57BL/6J mice (Center of Comparative Medicine Research, The University of Hong Kong) were maintained under a controlled environment (23 ± 1 ℃, 50–60%humidity, 12 h light/dark cycles) with foster mothers. All animal experimental procedures were approved by the Committee on the Use of Live Animals in Teaching and Research at the University of Hong Kong (CULATR 5544-20) and received humane care according to the criteria outlined in the “Guide for the Care and Use of Laboratory Animals. ” The mice were intraperitoneally injected with a single dose of diethylnitrosamine (DEN; 25 mg/kg) . At 6 weeks, the mice were provided with high-fat high-cholesterol (HFD) diet (containing 60%fat, 20%carbohydrates, 19.5%proteins, and 0.5%cholesterol, TP26304, trophic diet, Nantong, China) until sacrifice. At 12 weeks old, the mice were then randomly distributed and trained for one week for voluntary oral MediGel sucralose gel administration. After finishing the training, the control group (Con) and Prohep group (Pro) were given daily either 0.25 g of MediGel sucralose (ClearH2O, ME, US) , which served as a control gel, or 7 × 109 CFU per mice of Prohep in a control gel from until sacrifice. Sorafenib group (Sora) was the positive control and was supplemented (30mg/kg) starting from week 24. In ProSora group, daily Sorafenib (30mg/kg) was given in combination with daily Prohep (7 × 109 CFU per mouse) from the age of 24 weeks until sacrifice. In PrePro-Sora group, mice received daily Prohep for four weeks from the age of 20 weeks before the combined usage of Prohep and Sorafenib until sacrifice. The supplementation of Sorafenib and Prohep were given in the morning (10 AM) and afternoon (3 PM) respectively.
Histopathological Analysis
Fresh mouse liver tissues were processed and embedded in paraffin or directly embedded with an optimal cutting temperature compound to make frozen sections (Sakura Finetek USA, lnc., Torrance) . The paraffin sections were prepared in 5 μm thickness, subsequently deparaffinized, and rehydrated in xylene and graded ethanol. The sections were stained with a hematoxylin and eosin (H&E) kit (BASO, Wuhan, China) according to the manufacturer's instructions to evaluate the morphology and hepatic pathology. The MASLD activity score (NAS) was used to evaluate steatosis, lobular inflammation, and hepatocyte ballooning. Based on the pathological appearance, steatosis, lobular inflammation, and hepatocyte ballooning were assessed as follows. For steatosis, 0: steatosis area < 5%; 1: steatosis area = 5–33%; 2: steatosis area = 33–66%; 3: steatosis area > 66%. For lobular inflammation, 0: no foci; 1: <2 foci per 200× field; 2: 2–4 foci per 200× field; 3: >4 foci per 200× field. For hepatocyte ballooning, 0: none; 1: few balloon cells; 1: many balloon cells. Sirius red staining (Abcam, Cambridge, UK) was performed to evaluate the degree of liver collagen deposition and fibrosis, and the degree of liver fibrosis was quantified by calculating the percentage of the positive red staining area over the total colon area measured. Oil red O (ORO) staining (BASO, Wuhan, China) was used to evaluate the intrahepatic lipids. All sections were visualized with a biological microscope (Olympus, BX41) , and images were captured with a color digital camera (Olympus, DP72) .
Biochemical Measurements in Serum
A volume of 20 μL serum was diluted with deionized H2O for biochemical measurements. Total cholesterol, low-density lipoprotein cholesterol (LDL-C) , high-density lipoprotein cholesterol (HDL-C) , alanine aminotransferase (ALT) , and aspartate aminotransferase (AST) were measured using a cobas c111 analyzer (Roche Diagnostics; Indianapolis, IN, USA) according to the manufacturer’s instructions. Serum leptin was measured using Mouse/Rat Leptin Quantikine ELISA Kit (R&D Systems, Inc., USA) according to the manufacturer’s instructions.
Hepatic Content Extraction and Measurement
The hepatic total cholesterol (TC) , triglycerides (TG) , and glycogen were extracted and measured using a cholesterol assay kit HDL and LDL/VLDL (ab65390) , triglyceride assay kit quantification (ab65336) , and a glycogen assay kit (ab65620) . The absorbances were measured using a microplate reader at OD 570 nm for the colorimetric assay according to the manufacturer’s instructions (Abcam, UK) . For hepatic cytokines, liver tissues were homogenized with a RIPA buffer, and the levels of tumor necrosis factor-α (TNF-α) , interleukin-17 (IL-17) , interleukin-6 (IL-6) , and interleukin-10 (IL-10) were determined via measuring the absorbance at 450 nm according to the manufacturer’s instructions (BioLegend, lnc., San Diego, CA) . Hepatic glutathione (GSH) and superoxide dismutase (SOD) were measured using glutathione assay kit (Cayman Chemical, USA) and superoxide dismutase (SOD) colorimetric activity kit (Thermo Fisher Scientific Inc., USA) according to the manufacturer’s instructions.
Quantitative Real-Time Polymerase Chain Reaction (RT-qPCR)
RNA was extracted from mouse livers using a RNeasy Plus mini kit (Qiagen, Germany) . The integrity of the RNA was confirmed through gel electrophoresis, and the concentrations were confirmed using Thermo Scientific NanoDrop 2000/2000c spectrophotometers. cDNA was synthesized from 2 μg of RNA, using a HiScript II QRT supermix for qPCR (Vazyme Biotech, Nanjing, China) . For RT-qPCR, a reaction mixture containing an AceQ qPCR SYBR Green Master mix (Vazyme Biotech, China) , cDNA and primers was prepared. The sequences of primers used in this study are listed in Table 2) . Relative gene expression was normalized to GAPDH and calculated by the 2–ΔΔCt method.
Western Blot Analysis
Protein extraction from liver tissues was performed using RIPA buffer supplemented with protease and phosphatase inhibitor from Sigma-Aldrich (St. Louis, MO, USA) . The extracted protein was centrifuged at 13,000× g for 15 minutes at 4℃. The total protein content was quantified using the DC protein assay from Bio-Rad (Hercules, CA, USA) . An equal amount of extracted protein was loaded onto 10%SDS-PAGE and transferred onto a polyvinylidene fluoride (PVDF) membrane. The membrane was blocked with 5%non-fat milk or BSA and incubated overnight at 4℃ with primary antibodies, including anti-FAS (1:1000) (ab271016) , PI3K signaling pathway panel (ab283852) , anti-FOXO3A (ab12162) , anti-Heme Oxygenase 1 antibody (HO-1) (ab13248) and anti-GAPDH mouse (ab9485) antibody from Abcam (Cambridge, UK) . After washing, the membrane was incubated with secondary antibodies conjugated with horseradish peroxidase (HRP) , including goat anti-rabbit IgG (H+L) HRP conjugate or goat anti-mouse IgG (H+L) HRP conjugate from Biorad (CA, USA) . Protein bands were visualized using enhanced chemiluminescence reagents from Bio-Rad (CA, USA) and imaged using the ChemiDox XRS+ imaging system from Bio-Rad (CA, USA) . The intensity of the bands was quantified using ImageJ software from NIH (Bethesda, MD, USA) , and normalization was performed with reference to the intensity of GAPDH [Zhang et al. (2024) ] .
Short-Chain Fatty Acid Analysis
Short-chain fatty acids (SCFAs) , including acetate, propionate, butyrate, and valerate levels, were quantified in fecal samples using a modified protocol. Briefly, approximately 50 mg of fecal samples were homogenized in a 0.005 M sodium hydroxide buffer containing an internal standard (10 μg/mL acetic acid-d4, Cambridge Isotope Laboratories, USA) and centrifuged at 13, 200 x g for 20 minutes at 4℃. The liquid remaining after centrifugation was combined with a solution containing 1-propanol and pyridine in a 3: 2 ratio (volume/volume) , along with propyl chloroformate. Following a one-hour incubation at 60℃, 0.5 mL of hexane was introduced, and the mixture was centrifuged at 2000 x g for 5 minutes. A volume of 0.4 mL of the upper layer was transferred to a glass vial for GC-MS analysis (Agilent 6890 N-5973 GC-MS, USA) under the conditions described by Zheng, X., et al., A targeted metabolomic protocol for short-chain fatty acids and branched-chain amino acids. Metabolomics, 2013.9 (4) : p. 818-827. A 1 μL sample of the derivative was injected into the GC-MS system using the split mode at a ratio of 10: 1. The electron energy used was -70 eV, and mass spectral data were collected in a full-scan mode within the m/z range of 30 to 600. The levels of SCFAs were determined by creating calibration curves based on the peak areas of acetate, propionate, butyrate, and valerate against acetic acid-d4.
Total Microbial DNA Extraction and Metagenomic Sequencing
DNA of the fecal samples was extracted using a QIAamp PowerFecal Pro DNA kit (Qiagen, Germany) according to the manufacturer’s instructions and stored at -80 ℃. The integrity of the DNA was confirmed through gel electrophoresis, and the purity and concentrations were checked using Thermo Scientific NanoDrop 2000/2000c spectrophotometers (Thermo Scientific, DE, USA) . After passing the quality check, the extracted samples were sent to Beijing Genomics Institute Hong Kong Co., Limited (BGI, Hong Kong) for sequencing using the DNBseq platform (BGI, Tianjin) . The raw pair-end reads generated from metagenomic shotgun sequencing were aligned to the mouse reference genome GRCm39 using BWA-MEM for host contamination removal. Adaptor regions, low-quality reads, and PCR duplicates were further filtered with an in-house script as previously described [Aron-Wisnewsky, J., et al., Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol, 2020.17 (5) : p. 279-297] .
Taxonomic Profiling
SOAPnuke was used to remove sequences that were of low quality or made of artificial adapter Bowtie2 was used to exclude the reads mapped to phix and mice contamination (GRCm39) genomes. Taxonomic profiles were obtained using kraken2 with the pre-built standard database including archaea, bacteria, viral, plasmid, human, UniVec_Core (released on 10/9/2023) using confidence level set with 0.1. The raw taxonomic profile was then imported to bracken for species-level re-estimation. Taxa either presented in less than 10%samples or relative abundance less than 0.01%were removed from the down-stream analysis. R package phyloseq was used to aggregate the raw abundance table into a species-level counts per million (CPM) table. R package vegan was then used to determine alpha and beta diversity. Kruskal-Wallis test following with Dunn’s test and PERMANOVA (using adonis2 from the R package vegan) were applied for alpha diversity and beta diversity, respectively, to assess the overall composition difference between groups. ANCOM-BC was used to compare the abundance of taxa between groups. Linear dis-criminant analysis (LDA) was also used to lessen the bias present in the various differential methods Differentially abundant taxa were those fulfilled specific requirements including Fold change > 2 or <1/2, LDA score (log10 transformed) > 2, and False Discovery Rate (FDR, adjusted p value generated by ANCOM-BC) < 0.05. After finding the differential abundant species for each pair-wise comparison, sparCC algorithm was applied to find the correlation between those species. Only the correlations with p value < 0.05 and absolute value of r > 0.7 were considered and visualized in the network.
Functional Profiling
Concatenated clean paired-end reads were input to HUMAnN3 (v3.7) in order to evaluate the abundance of MetaCyc gene families. Kyoto Encyclopaedia of Genes and Genomes (KEGG) ontology (KO) , KEGG module were obtained by utilizing the HUMAnN3 regroup table function. ANCOM-BC was used to assess the differential abundances (DA) of KO and the abundance of metabolic pathways. KO and pathway that met FDR < 0.05 were considered as differentially abundant.
Statistical Analysis
All metagenomic statistical analyses and visualizations were carried out in the R programming environment, unless otherwise noted. Spearman’s rank correlation, found in the function cor. test of the R package stat, was utilized for general correlation analysis. Aside from the metagenomics analysis as described above, another statistical analysis was performed with GraphPad Prism 8.0 (GraphPad Software, San Diego, CA, USA) unless specified. Experimental results are expressed as means ± standard deviation (SD) . Outliers were identified using Grubbs’ test. Comparisons of differences between two groups were analyzed with Student’s t-tests. Comparisons of differences in more than two groups were analyzed with one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test. P values of <0.05 were considered statistically significant. A false discovery rate (FDR) (Benjamini–Hochberg) was used to adjust the p-value. Adjusted p value < 0.05 was used as a cut-off unless otherwise specified.
Results
Prohep protected tumor development in DEN-HFD HCC mice model
To determine the anti-tumorigenic effect of the probiotics mixture in naturally occurring HCC, Prohep was tested in a mouse model that mimics the progression from MASLD to HCC. Mice were intraperitoneally injected with a single dose of DEN, were fed a HFD and were given the treatment as described in the feeding scheme (Fig. 1A) . No significant difference in body weight was observed between the control group and the Prohep group. However, upon Sorafenib treatment, up to 35%of weight loss in 6 weeks was witnessed in Sora group compared to Control group, while the addition of Prohep could not rescue the weight loss in Pro-Sora and PrePro-Sora groups.
After harvesting the organs, Pro, Pro-Sora and PrePro-Sora groups showed significantly retarded tumor development as evidenced by significant decreases in the liver/body weight ratio, surface tumor counts, and surface maximum tumor sizes compared to Control group. While Sora group is effective in reducing liver/body weight ratio and surface maximum tumor sizes but not surface tumor counts. Prohep showed potential adjuvant effect as it greatly improved the therapeutic effect of Sorafenib. Of note, the tumor counts were significantly lower in the ProSora and PrePro-Sora groups compared to Sorafenib alone. The tumor burden was further confirmed by analysis of H&E staining of mouse liver sections (Fig. 1B) . Additionally, a significant reduction in Ki-67 staining was observed in Prohep-treated mice compared to control, indicating a decrease in cell proliferation. Conversely, compared to Con group a significant increase in C-CASP3 staining was observed in Prohep-treated mice liver, suggesting an increase in cell apoptosis, which plays a crucial role in the treatment of liver cancer by promoting targeted cell death. Furthermore, through histopathological examination of lung sections, a reduction in lung metastasis was found in the Prohep group when compared to Con group.
Table 4: Prohep treatment reduce the mean maximum tumor size
The mean maximum tumor size in Con, Pro, Sora, Pro-Sora and PrePro-Sora groups are 0.78, 0.23, 0.3, 0.26, 0.16 (cm) respectively. Prohep treatment reduced the mean maximum tumor size by 70.51%compared with the control. While the old formula of Prohep showed reduction of the tumor size by 40%compared with the control.
Prohep ameliorated MASLD progression in DEN-HFD HCC mice model
In addition to reducing the HCC incidence, Prohep also impeded the progression of MASLD in DEN-HFD HCC mice model. The anti-tumor effects were accompanied by ameliorated MASH progression, as evidenced by lower hepatic steatosis, fibrosis area, and NAS score (Fig. 2) . Hepatic inflammation, such as macrophage infiltration revealed by F4/80 IHC staining (Fig. 2) and reduced pro-inflammatory cytokines TNF-α, IL-17, IL-1β IL-6 were also ameliorated in the Prohep-treated group. The improvement of liver health was secondly shown in reduced serum levels of ALT, AST, cholesterol and leptin. These findings suggested that Prohep is beneficial and protective against MASLD progression.
Low dose Sorafenib treatment ameliorated the major hallmarks of MASLD including hepatic steatosis, inflammation and fibrosis but liver markers such as cholesterol, ALT and AST, and leptin in serum. Noteworthily, Pro-Sora and PrePro-Sora have significantly reduced TNF-α level and less macrophage infiltration as indicated by F4/80 staining that could lead to NAS score decrease when compared to Sora group. Beyond that, PrePro-Sora could reduce the serum leptin level compared to Sora group, indicating a remission from leptin resistance.
Prohep modulated cancer metabolism in tumor microenvironment via activating AMPK signaling pathway
High oxidative stress is often observed in the tumor microenvironment. Compared to control group, Prohep treated mice had higher hepatic SOD and GSH levels that could lead to less oxidative stress in the tumor microenvironment. Meanwhile, excessive lipid accumulation is another hallmark in MASLD-HCC tumor microenvironment. Upon Prohep treatment, both liver total cholesterol and triglyceride levels significantly decreased compared to Con. At the same time, the genes responsible for lipogenesis and lipid uptake, FAS, ACC1, and CD36 had decreased expressions in Pro group compared to Con group. Its underlying mechanism was further investigated and found a distinct activation of AMPK signaling pathway in Pro group when compared to the Con group, Sora group, and Pro-Sora in liver IHC staining (Fig. 3A) . The decreased expression of FAS protein and increased oxidative stress-related protein, HO-1 and FOXO3A were further confirmed in western blot (Fig. 3B) .
Sora group also showed significantly reduced FAS, ACC1 expression accompanied with decreased liver total cholesterol and triglyceride levels when compared to Con group. It could be linked to the body and liver weight drop after Sorafenib treatment. Compared to Sora group, PrePro-Sora showed a significantly increased hepatic SOD level that could help reduce the oxidative stress in the tumor microenvironment.
Prohep suppressed the activity of the PI3K/mTOR pathway in DEN-HFD induced HCC mouse models
PI3K/mTOR pathway is responsible for downstream signals that promote cancer cell growth and proliferation, making it a crucial target for HCC treatment. In the Pro group, the protein expression of phosphorylated PI3K and mTOR were significantly reduced, leading to inhibited cancer proliferation signaling (Fig. 4) . Even though the decrease of p-PI3K and mTOR were not significantly lower in Sora group, phosphorylated PI3K level was significantly suppressed in PrePro group that could contribute to anti-tumor effect.
Prohep treatment significantly altered the gut microbiome
The gut microbiota compositions of all groups were investigated. Notably altered microbial composition was observed in Sora, Pro, Pro+Sora, PrePro+Sora compared to Con group as illustrated in the PCoA plot (Table 1) . Notably, PrePro-Sora group showed significant differentiation from Sora group in gut microbiota composition while Pro-Sora group did not (Table 1) , indicating the modulation effects of giving Prohep in advance. The analysis of alpha diversity did not reveal any notable differences among the groups studied. This observation is consistent with the results of a recent study of MSALD-HCC gut microbiota. It found that HFD feeding significantly reduced alpha diversity after 2 weeks compared to normal chow feeding. However, there was no significant difference in alpha diversity between the HFD and normal chow feeding groups after a longer period of 16 weeks or 14 months.
Differential bacteria between Con and Pro groups was first investigated. 20 genus and 27 species were enriched in Con group, while 19 genus and 53 species were enriched in Pro group. The signature differential genera and species were presented. In Prohep group, naturally the species from Prohep, Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus rhamnosus, Lactobacillus plantarum, Streptococcus thermophilus, Bifidobacterium breve, Lactobacillus helveticus, Bifidobacterium animalis ssp. lactis showed significantly higher abundance due to the supplementation. Secondly, several well-studied probiotics species Akkermansia muciniphila, Parabacteroides distasonis, Phocaeicola vulgatus and Alistipes senegalensis, Phocaeicola sartorii (previously named as Bacteroides sartorii) also showed increase in Pro group. Thirdly, certain pathogenic pecies, Faecalibaculum rodentium, Clostridioides difficile, Leptogranulimonas caceicola, Eggerthella lenta, Clostrium perfringens were shown to enriched in control group but deceased in Pro-group. Interestingly, correlations were identified between probiotics and pathogenic bacteria in Prohep-treated mice. As indicated by the green line, the abundance of S. thermophilus against C. perfringens, L. plantarum and L. rhamnosus against L. caecicola, C. perfringens, Thomasclavelia ramose and Thomasclavelia spiroformis, B. animalis against C. perfringens, P. distasonis inhibit the growth of E. lenta and F. rodentium, L. acidophilus against T. ramose, T. spiroformis and C. perfringens, A. muciniphila against F. rodentium and so on, implying that Prohep-enriched probiotics may antagonize pathogenic bacteria in HCC. Moreover, some of the gut microbiome signatures from Pro-group were significantly correlated with metabolic indices. In addition to the abundance of the constituent species in Prohep, L. paracasei, B. breve, L. plantarum, L. rhamnosus, L. acidophilus, B. animalis ssp. lactis, S. thermophilus and L. helveticus, the abundance of P. vulgatus, P. sartorii, mucispirillum schaedleri were also negatively correlated with the tumor counts and maximum tumor size. Among the newly identified ones, M. schaedleri and P. vulgatus’ abundances were positively associated with many probiotics species, including Prohep components. Taken together, these data suggest that Prohep might reverse microbial dysbiosis in MASLD-HCC.
Table 1. Beta_adnois_pairwise analysis
Table 2: Primer sequence of RT-qPCR
Prohep increased gut microbiota–derived propionate and valerate
To characterize the functional pathway of Prohep, a significant differential Metacyc pathway profile was plotted. The pathway “pyruvate fermentation to propanoate I” , which lead to the production of propionic acid was found to be significantly higher in mice treated with Prohep (Fig. 5A) . Three enzymes, fumarate reductase (1.3.5.4) , methylmalonyl-CoA carboxyltransferase (2.1.3.1) and malate dehydrogenase (1.1.1.37) participated and the synthesis pathways were significantly elevated (Fig. 5A-5C) . In the contrary, the fatty acid β-oxidation pathways that mediated its degradation were lower in Prohep group. The metagenomic predicted functions were in consistency with the increased fecal propionic acid concentration in Pro group (Fig. 5D) . We’ ve further investigated the production of other types of SCFAs. Apart from propionic acid, the production of valeric acid was also elevated in Prohep group (Fig. 5D) . Further investigation revealed that Bifidobacterium breve, Bifidobacterium animalis ssp. lactis and Phocaeicola sartorii were significantly positively linked with both propionic acid and valeric acid, Parabacteroides distasonis and Lactobacillus acidophilus were positively associated with valeric acid.
Prohep improve the metagenomic profiling in low dose Sorafenib treated HCC mice
The beneficial effect of applying Prohep have also been explored in combination with Sorafenib. In addition to the increased abundance of Prohep components, L. helveticus, L. plantarum, L. rhamnosus, L. paracasei, L. acidophilus, B. breve, B. lactis and S. thermophilus, the abundance of T. spiroformis, T. ramose, and Romboutsia ilealis were decreased in Pro-Sora and PrePro-Sora group. Then, the species correlations among groups was investigated. With the addition of Prohep, more interactions among species were spotted in PrePro-Sora (854 correlations) than in Sora (720 correlations) and Pro-Sora (638 correlations) groups. More importantly, PrePro-Sora (296 negative correlations) and Pro-Sora (206 negative correlations) groups had more negative correlations compared to Sora (150 negative correlations) . And several probiotics species contributed to the inhibition of certain known pathogenic species, say, B. breve, L. acidophilus, L. paracasei, L. rhamnosus, L. plantarum, L. acidophilus, S. thermophilus against Bacteroides faecium. Interestingly, the production of butyric acid were promoted in both Pro-Sora and PrePro-Sora groups. Increased butyric acid was positively correlated with the abundance of Brachyspira murdochii, Brachyspira hydodysenteriae, Brachyspira hampsonii, L. helveticus, L. acidophilus, B. breve, B. animalis ssp. lactis. The Prohep components, L. helveticus, L. plantarum, L. rhamnosus, L. paracasei, L. acidophilus, B. breve, B. lactis and S. thermophilus, were identified to be negatively correlated with tumor counts and tumor maximum size again in the correlation analysis includes only Sora, Pro-Sora, and PrePro-Sora group. tumor counts and tumor maximum size were negatively correlated with Helicobacter typhlonius and Helicobacter apodemus as well.
Discussion
The significant role of gut microbiota in the pathogenesis of MASLD-HCC has been increasingly recognized. Several studies have highlighted the occurrence of gut dysbiosis in various stages of chronic liver diseases, ranging from MASLD to cirrhosis and HCC. Additionally, gut microbiota has implications for the pharmacokinetics of cancer drugs and the management of cancer treatment. In this study, the anti-tumorigenic effect of Prohep was demonstrated and the adjuvant effect of Prohep to low dose-Sorafenib treatment in DEN-HFD induced MASLD-HCC mouse model. Additionally, the current study was the first to explore the adjuvant effect of probiotics on low dose Sorafenib, which considers both the treatment efficacy and side effect management.
As Prohep is orally administered and colonizes the gut, the crosstalk between Prohep supplementation and gut microbiota during NAFLD-HCC formation was investigated. Metagenomic profiling revealed that Prohep modulated gut microbiota dysbiosis, contributing to the inhibition of NAFLD-HCC. Concomitant with the improvement of gut dysbiosis, alterations in gut microbiota signatures were observed to alleviate HCC etiology. Notably, the growth of several probiotic species was observed to increase following Prohep treatment. Akkermansia, a newly identified anticancer bacterium, has been shown to inhibit obesity and promote intestinal barrier function while delaying cancer progression. Multiple studies have linked the decreased abundance of Akkermansia and A. muciniphila in NASH-HCC patients compared to NASH-cirrhosis patients without HCC. M. intestinale was found to be enriched in a blueberry malvidin-3-galactoside-treated HepG2 cell-injected tumor model and identified as an important factor affecting the microbial tricarboxylic acid (TCA) cycle KEGG pathway. Additionally, the significantly elevated species P. vulgatus was shown to mitigate the development of MASLD by promoting the production of metabolite 3-Hydroxyphenylacetic acid (3-HPAA) , which restrained lipid accumulation in liver cells. Furthermore, altered gut microbiota was found to antagonize pathogenic bacteria in MASLD-HCC, with the growth of pathogenic species such as T. ramose, enriched in MASH and HCC patients with alcohol-related liver disease who develop HCC, being significantly suppressed in the Prohep group. Similarly, D. piger, whose increase leads to intestinal permeability and overexpression of CD36 in the liver, thereby aggravating MASLD, was also found to be suppressed in the Prohep group. In addition, specific bacteria such as, Gordonibacter pamelaeae P7-E3, Eggerthella lenta P7-G7, Adlercreutzia equolifaciens P11-C8, which showed decreased abundance in Pro group, were identified to be top producers of converting LCA into 3-oxoLCA, which further suppressed Th17 cell differentiation. Many experimental tumor models in mice have provided evidence of favoring Th17 cells in the control of cancer growth. Some other species like, Hungatella hathewayi was established as colorectal cancer (CRC) -promoting bacteria as it could induce DNA methyltransferase to tumor suppressor genes promoters in host cells. C. difficile, which bears responsibility for gastrointestinal infections and is linked to colorectal cancer. E. lenta has been reported to be enriched in multiple acute and chronic inflammatory diseases. F. rodentium, originated from genus Faecalibaculum, is well studied for being proinflammatory bacteria that may impair the gut barrier as well.
Further analysis of the metagenomic data revealed a significant increase in the "pyruvate fermentation to propanoate I" pathway in the Prohep group. Propanoate, also known as propionate, is a short-chain fatty acid (SCFA) that is a final product of dietary fiber fermentation by anaerobic bacteria. SCFAs, including acetate, propionate, butyrate, and valerate, are known to exert multiple beneficial effects on human lipid and glucose metabolism. To verify the function prediction of the metagenomic data, targeted metabolomic analysis was performed, which confirmed the significant upregulation of SCFAs, specifically propionate and valerate, in the Prohep group. Their production was positively correlated with SCFA-producing lactic acid bacteria (LAB) species in Prohep, as well as with P. sartorii and P. distasonis. Both P. sartorii and P. distasonis are known to be susceptible gut-indigenous bacteria that respond to the ingestion of functional food ingredients, resulting in synergistic effects on the host. Propionate has been demonstrated to act as an inhibitor of HCC proliferation by activating the GPR43 signaling pathway. Furthermore, a significant reduction in valerate levels in mice serum was observed in the HCC group compared to the healthy group. In a recent study, L. acidophilus supplementation in MASLD-HCC mice was found to exhibit anti-tumorigenic effects by secreting valerate. Both propionate and valerate have been found to possess histone deacetylases (HDAC) -inhibitory activity, which can release the suppression of apoptosis key effector CASP3 activity from HDACs and induce cancer cell apoptosis. The results disclosed herein are aligned with these findings, as higher levels of propionate and valerate and increased CASP3 activity were observed in the Prohep group, potentially contributing to tumor inhibition.
Modulating the tumor microenvironment is a strategy for targeting HCC. Propionate has previously been shown to regulate hepatic glucose and lipid metabolism via AMPK activation in human HepG2 hepatocytes. In the current study, it was identified that liver AMPK was activated in the Prohep group. At the same time, increased propionate level has been linked to the activation of liver AMPK. Liver cancers are characterized by upregulation of lipid catabolism and oxidative stress. AMPK, as a primary regulator of cellular energy homeostasis, often contributes to metabolic-associated disorders and is commonly found to be dysfunctional in patients with HCC. Low AMPK activation is correlated with aggressive clinicopathologic features and poor prognosis. In liver disease, activated AMPK can inhibit the synthesis of fatty acids and cholesterol by downregulating the expression of lipogenesis genes such as FAS and ACC. AMPK activation has also been reported as an inducer of CD36 expression, leading to lipid accumulation in hepatocytes. Upon AMPK activation, decreased expression of FAS, ACC, and CD36 was observed in the Prohep group. Activation of the AMPK signaling pathway has been shown to contribute to the regulation of reactive oxygen species (ROS) . AMPK regulates FOXO3A and HO-1, downstream target genes, in response to cellular oxidative stress. Both FOXO3A and HO-1 contribute to the prevention of the increase of oxidative stress. Metformin supplementation has been reported to inhibit the development of hepatocellular carcinoma via FOXO3 activation through the AMPK pathway. Furthermore, increased SOD and GSH activity helped to relieve oxidative stress due to hepatic fatty acid overload. Overall, the decrease in fatty acid uptake, synthesis of lipids and oxidative stress damage hindered the lipotoxicity-driven pathogenesis of metabolic diseases and lowered the risk of HCC.
The pro-inflammatory and pro-fibrotic liver microenvironment promotes hepatocarcinogenesis. As a result of Prohep supplementation, inflammation in the liver, as represented by the macrophage marker F4/80 and expression levels of TNF-α, IL-17, IL-6, and IL-1β, were significantly reduced. Meanwhile, fibrosis scars were also reduced after Prohep supplementation. The multiple-hit hypothesis of the pathogenesis of MASH and subsequent HCC development suggests that multiple hepatotoxic insults occurring in parallel contribute to disease progression.
Oncogenic signaling transduction pathways, such as the phosphoinositide 3-kinase (PI3K) , AKT, and mammalian target of rapamycin (mTOR) pathways, which play a major role in regulating cell proliferation, survival, and angiogenesis, are of great importance in cancer therapy. Dysregulation of the PI3K/AKT/mTOR signaling pathway is common in HCC. Previous studies have demonstrated that sodium propionate supplementation suppressed the PI3K/Akt/mTOR signaling pathway and attenuated LPS-induced epithelial-mesenchymal transition. In the current study, PI3K/mTOR signaling was suppressed in the Prohep group, leading to inhibited cancer cell proliferation.
Recent studies have shown that the gut microbiome and its metabolites can enhance the efficacy of anti-tumor drugs from various perspectives. In a preliminary study, it was found that Prohep supplementation for 4 weeks significantly altered the gut microbiota's alpha and beta-diversity. Combining a low dose of Sorafenib with Prohep, especially when Prohep was given 4 weeks ahead, effectively reduced surface tumor counts, improved NAS activity, and decreased F4/80 and TNF-α expressions. Hepatic inflammatory microenvironments contribute to Sorafenib resistance through anti-apoptotic protein Mcl-1 from Bcl-2 family mediated by STAT3. Elevated expression of both STAT3 and BCL2 have been linked to Sorafenib resistance. With Prohep supplementation, liver inflammation was alleviated. Decreased expression of hepatic STAT3 and relatively lower expression of BCL-2 induced by additional supplementation of Prohep could help enhance the treatment efficacy of Sorafenib. Moreover, in the metabolomic analysis, “Palmitate biosynthesis” was enriched in Pro-Sora and PrePro-Sora groups. Methyl palmitate was once studied in HCC cells as a suitable adjuvant for Sorafenib therapy that could help reduce in vivo toxicity and enhance anti-cancer effects. Further investigation revealed that gut microbiome alteration led to increased butyrate production. Previous studies have shown that butyrate supplementation improves the therapy efficacy of Sorafenib. Butyrate can enhance T-cell immunity and potentially improve the response to targeted drug therapy in mice orthotopic HCC models. Therefore, the synergistic interaction among the components can amplify the therapeutic effect, offering benefits such as increased efficiency and reduced toxicity.
In conclusion, the results presented herein suggest that Prohep supplementation effectively inhibits the development of MASLD-HCC, as demonstrated by a reduced tumor count, and maximum tumor size compared to the control group. Proheps were found to alleviate crucial factors of MSALD-HCC development, such as steatosis, fibrosis, and inflammation. A sharp decrease in the proliferation marker Ki67 was also observed across all treatment groups. Prohep supplementation led to the activation of AMPK, which plays an important role in cellular and systemic energy homeostasis. Moreover, Prohep group inhibited PI3K/mTOR pathway, a cancer proliferation pathway. The therapeutic effect of Prohep can be partially attributed to the modulation of gut microbiota and the increased production of propionate and valerate. Prohep modulated not only the diseased gut microbiome but also the Sorafenib-treated gut microbiome, leading to increased SCFA, butyrate production, hence improving treatment outcomes. Additional supplementation of Prohep to low-dose Sorafenib significantly reduced tumor counts in MASLD-HCC model while low-dose sorafenib alone didn’ t. The potentiating effects could be attributed partially to ameliorated liver inflammation and increased antioxidative activity. The current findings demonstrate the prophylactic potential of Prohep alone and adjuvant effect on low dose sorafenib.
Example 2: Prohep attenuates colorectal tumorigenesis in murine AOM/DSS model by suppressing STAT3, inducing apoptotic p53 and modulating gut microbiota
Materials and methods
Chemicals and antibodies
Azoxymethane (AOM) and dextran sulfate sodium (DSS) were purchased from Sigma-Aldrich (St. Louis, MO, USA) and TdB Labs (Uppsala, Sweden) respectively. Prohep probiotic mixture formula composed of Lactobacillus helveticus, Bifidobacterium lactis, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus paracasei , Lactobacillus acidophilus, Bifidobacterium breve and Streptococcus thermophilus. Prohep was produced in lyophilized powder under GMP (Fukopharma, Finland) . All primary antibodies were bought from Abcam (Cambridge, UK) and CST (Massachusetts, USA) while the secondary antibodies were purchased from Bio-Rad (Hercules, CA, USA) .
Animals and experimental setup
The experimental animals were obtained from the Centre for Comparative Medicine Research (CCMR, HKU) . 6 weeks old male BALB/c mice were placed in 12-hour light/dark cycle with a normal chow diet and drinking water given ad libitum for acclimatization upon the first week of receival. Voluntary gel administration training was carried out for the week afterwards. The chow diet being was taken away overnight on the first day and 0.20 g of Sucralose gel (Maine, USA) was given the next morning in individual cages. Mice were allowed to return to their house cages after consuming the given gel. The training continued for another two days but chow diet intake was not constrained. Mice group allocation was as follows: 1. Healthy control (H) , 2. AOM/DSS control (AOMDSS) , 3.5-FU (F) , 4. Prohep (P) and 5. Prohep+5-FU (PF) . As previously described, the AOM/DSS model was established by injecting the mice with AOM (10mg/kg) intraperitoneally at the start of the experiment while PBS was injected for the healthy control group. One week of DSS (2.5%) , added in drinking water, was provided and be replaced by regular drink water in the next week. Three cycles of DSS treatment were given in total. Healthy control group received regular drinking water for all time. Prohep administration started at week 0. Prohep (7 × 109 CFU per mice) were infused intoSucralose gel (Maine, USA) and provided to each mouse of Prohep and Prohep+5-FU groups at 0.20 g every other day until the end of the experiment. The dosage of Prohep was based on the previous studies. The other groups receivedSucralose gel without modification as control. Starting at week 5, 5-FU (35 mg/kg) was injected intraperitoneally to 5-FU and Prohep+5-FU groups weekly. PBS (vehicle) was injected into other groups. At week 12, the animals were sacrificed, and the colon was cut open longitudinally to determine the tumor count and colon length.
Colon samples were frozen at -80℃ for biochemical analysis and were fixed in 10%formalin for histology analysis. Fecal samples were collected and frozen at -80℃ on the sacrifice day. Liver samples were weighted. All protocols and procedures were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong (CULATR No. 5584-20) .
Colon histological analysis
Formalin-fixed colon samples were embedded into paraffin blocks. Specimens were sectioned on slides and deparaffinized to undergo staining procedures. Haematoxylin and eosin (H&E) staining was performed according to the manufacturer’s manual (BASO, Wuhan, China) . Adenoma counts were performed with light microscopy. Sirius red staining was carried out according to the manufacturer’s manual (Abcam, Cambridge, UK) . The percentage of positive red staining area over the total colon area measured was used to quantify the extent of colonic collagen fibrosis.
Immunohistochemistry (IHC) staining
IHC staining was performed with heat-induced antigen retrieval method with sodium citrate (pH6) or Tris-EDTA (pH9) . Endogenous peroxidase activity was inhibited with 3%H2O2 and sections were blocked with CAS-block reagent (Invitrogen, Waltham, MA, USA) for one hour. Endogenous peroxidase activity was inhibited with 3%H2O2 and blocked with CAS-block reagent (Invitrogen, Waltham, MA, USA) for one hour. Primary antibodies incubation (1: 100) was performed at 4℃ overnight followed by secondary antibodies incubation (1: 250) at room temperature for one hour. DAB (Abcam, Cambridge, UK) chromogen reaction and haematoxylin counterstaining were performed to visualize the positively stained area. The percentage of positively stained area was determined with ImageJ software (NIH, USA) and histoscore was calculated by multiplying the percentage with the intensity of staining graded from 0, non-stained; 1, weakly stained; 2, moderately stained; and 3, strongly stained.
Cytokines ELISA analysis
Protein extraction was performed by homogenizing colonic samples in RIPA buffer with protease and phosphatase inhibitor (Sigma-Aldrich, St. Louis, MO, USA) and protein samples were collected by centrifuging the homogenate. The total protein content was measured using DC protein assay (Bio-Rad, CA, USA) . ELISA analysis of TNF-α was performed using Mouse ELISA MAXTM Set (BioLegend, CA, USA) according to the instructions of manufacturer. The absorbance was measured using SpectraMax iD3 microplate readers (Molecular devices, CA, USA) .
Western blot analysis
Diluted protein was electrophorized in 10%SDS-PAGE gel and transferred to a polyvinylidene fluoride (PVDF) membrane. The membrane was blocked with 5%non-fat milk or BSA, followed by overnight exposure of anti-p53 (1: 1000) (Abcam, Cambridge, UK) at 4℃. Secondary antibody exposure was performed the next day with goat anti-rabbit IgG (H+L) HRP conjugate or goat anti-mouse IgG (H+L) HRP conjugate (1: 4000, Bio-Rad, CA, USA) for one hour at room temperature. Protein bands were visualized with enhanced chemiluminescence reagents (Bio-Rad, CA, USA) using ChemiDox XRS+ imaging system (Bio-Rad, CA, USA) .
Metagenomics
Fecal samples obtained on the sacrifice date were used to extract microbial DNA usingPro DNA kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. Extracted DNA was sent to BGI Genomics (Shenzhen, China) for whole genome shotgun sequencing. SOAPnuke was used to filter out adaptor reads that were of low quality or artificial. Bowtie2 was used to exclude the reads mapped to phix and mice contamination (GRCm39) genomes. Using the most recent release (2023-05-10) of the NCBI nr database, which contains archaea, bacteria, viruses, fungi, and microbial eukaryotes, taxonomic profiles were produced using Kaiju with parameter "-e 5" . Taxa excluded from the downstream analysis were those that were either present in less than 10%of the samples or had a relative abundance of less than 0.01%. R package phyloseq was used to aggregate the raw abundance table into a species-level counts per million (CPM) table. R package vegan was then used to determine alpha and beta diversity. Dunn test and PERMANOVA (using adonis2 from the R package vegan) were applied for alpha diversity and beta diversity, respectively, to assess the overall composition difference between groups. ANCOM-BC was used to compare the abundance of microorganisms between AOM/DSS with and without treatment. Linear discriminant analysis (LDA) was also used to lessen the bias present in the various differential methods. Microorganisms that met certain criteria were deemed differentially abundant, including Fold change ≥ 2 or ≤ 1/2, LDA score (1og10 transformed) ≥ 2, and FDR ≤ 0.05. Concatenated paired-end read files were input to HUMAnN3 (v3.7) in order to evaluate the abundance of metabolic Metacyc pathways and gene families abundance. Kyoto Encyclopedia of Genes and Genomes (KEGG) ontology (KO) , KEGG module, and Metacyc pathway profiles were obtained by utilizing the HUMAnN3 regroup table function. ANCOM-BC was used to assess the differential abundances (DA) of KO and the abundance of metabolic pathways. KO and pathways that met FDR value < 0.1 were deemed differentially abundant. Dunn’s post hoc test was employed after the Kruskal-Wallis test for general statistical comparisons between several groups. Spearman’s rank correlation, found in the function cor. test of the R package stat, was utilized for general correlation analysis. The p-value was adjusted for multiple testing adjustments using a false discovery rate (FDR) (Benjamini–Hochberg) . All statistical analyses and visualizations were carried out in the R environment, unless otherwise noted.
Short-chain fatty acids (SCFAs) analysis on fecal content
Fecal SCFAs content was determined using gas chromatography-mass spectrometry (GC-MS) according to previous studies. In short, fecal samples were homogenized in in 0.005 M sodium hydroxide with internal standard (10 μg/mL acetic acid-d4) and centrifuged at 13, 200 × g for 20 mins. The supernatant was then mixed with 0.5mL of 1-propanol/pyridine (3: 2, v/v) and 0.1mL of propyl chloroformate. The SCFAs were derivatized by vertexing the mixture for 1 minute and incubating it at 60℃. 0.5mL hexane was then added, vortexed, and centrifuged at 2000 × g for 5 minutes. 400μL of the top layer was used for GC-MS analysis (Agilent 6890 N-5973 GC-MS, USA) with conditions based on previous studies. By constructing the calibration curves using the response ratios of acetic acid, propionic acid, and butyric acids against acetic acid-d4, the concentration of the SCFAs was obtained.
Statistical analysis
Statistical analysis was performed with GraphPad Prism 8.0 (GraphPad Software, San Diego, CA, USA) . All data were presented as mean ± standard deviation (SD) . Student’s t-tests or Mann–Whitney U tests were adopted to compare differences between two groups. One-way analysis of variance (ANOVA) followed by Tukey’s multiple comparisons test or the Kruskal–Wallis test followed by Dunn’s multiple comparisons test was computed to compare differences in more than two groups. The Spearman correlation coefficient was employed to compute the correlation between two variables. For multiple-testing corrections, a false discovery rate (FDR) (Benjamini–Hochberg) was used to adjust the p value. A p-value < 0.05 was recognized as having statistical significance.
Results
Prohep significantly improved survival and reduced colorectal tumorigenesis of AOM/DSS mice
Among all treatment groups, only Prohep significantly increased the survival rate against the AOM/DSS group (p < 0.05) (Fig. 6A) . While 5-FU, Prohep and Prohep+5-FU all significantly reduced the total tumor count induced by AOM/DSS (p < 0.05) , only Prohep group also significantly reduced the total tumor size (p < 0.05) (Figs. 6B-6D) . The caecum weight was elevated in AOM/DSS group (p < 0.05) . No significant difference in caecum weight between the Prohep group and the healthy control group (p > 0.05) was found, indicating that Prohep were effective in preventing caecum enlargement (Fig. 6E) . Similar to the 5-FU group, Prohep+5-FU did not improve survivability and reduce total tumor size as seen from the Prohep group. In addition, the administration of 5-FU led to the reduction of liver weight in 5-FU and Prohep+5-FU groups when compared with healthy and Prohep groups (p < 0.05) (Fig. 6F) .
In terms of histopathology, the elevated colonic crypt depth was reduced by Prohep (p < 0.05) , but not 5-FU and Prohep+5-FU (Fig. 7A, Fig. 7E) . Meanwhile, both hyperplasia and inflammation scores were significantly reduced by Prohep and Prohep+5-FU correspondingly (p < 0.05) (Fig. 7B-7C, Fig. 7E) . The reduction effects were not observed in the 5-FU group. From Sirius red staining, Prohep is the only group which alleviated the heightened Sirius red positive percentage (p < 0.05) , which 5-FU and Prohep+5-FU could not (Fig. 7D, Fig. 7F) . It indicated that Prohep as the only treatment group to reduce the degree of collagen fibrosis in colon. The proliferative and apoptotic status was evaluated with IHC staining. Prohep and Prohep+5-FU significantly reduced the H-score of the proliferative marker, Ki67 (p < 0.05) , but 5-FU did not (Fig. 8B, Fig. 8I) . On the other hand, 5-FU and Prohep improved the H-score of the apoptotic marker, c-casp3 (p < 0.05) , but not by Prohep+5-FU (Fig. 8F, Fig. 8I) .
Prohep inhibited colorectal tumorigenesis through suppressing proliferative p-STAT3 and c-jun/c-fos and promoting apoptotic p53
AOM/DSS elevated the levels of pro-inflammatory cytokine, TNF-α (p < 0.05) (Fig. 8A) . All treatments including 5-FU, Prohep and Prohep+5-FU significantly reduced the concentration of TNF-α (p < 0.05) (Fig. 8A) . Prohep significantly suppressed the level of proliferative p-STAT3, c-jun and c-fos (p < 0.05) (Fig. 8C-8E, Fig. 8I) . No suppressive effects on cancer cell progression were observed in the 5-FU and Prohep+5-FU groups. Although 5-FU also demonstrated elevation in the apoptotic marker, c-casp3 (p < 0.05) (Fig. 8F, Fig. 8I) , only Prohep could significantly promoted the protein level of apoptosis inducing p53 (p < 0.05) (Figs. 8G-8H) . No alteration of apoptotic markers was observed from the Prohep+5-FU group. It was thus suggested that Prohep suppressed STAT3, c-jun and c-fos and activate p53 to inhibit colorectal tumorigenesis.
Prohep modified gut microbiota and promote biomarkers to elevate beneficial metabolites biosynthesis and acetate concentration
The administration of Prohep significantly modulated the abundance of gut microbiota compared to AOM/DSS. Prohep elevated the abundance of Ligilactobacillus ruminis, Ligilactobacillus murinus, Adlercreutzia caecimuris, Ligilactobacillus animalis, Adlercreutzia mucosicola, Enterococcus faecalis, Sangeribacter muris, Muribaculaceae bacterium Isolate-001 (NCI) , Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila, Muribaculaceae bacterium Isolate-080 (Janvier) , Duncaiella dubosii, Prevotella sp. PTAC, Muribaculaceae bacterium Isolate-007 (NCI) , and Helicobacter typhlonius (p < 0.05) . Besides, Prohep lowered Clostridum sp. CAG: 510, bacterium 0.1xD8-71, Candidatus Gastranaerophilus sp. (ex Termes propinquus) , Vermiculatibacterium agrestimuris, Roseburia sp. CAG: 309 (p <0.05) (Fig. 9A) .
From the identified bacteria, Spearman’s correlation was carried out to evaluate the inhibitory effects on colorectal tumorigenesis by modulating these bacteria. Prohep elevated Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus and Candidatus Borkfalkia ceftriaxoniphila were found to be inversely correlated to the total tumor count (p <0.05) (Fig. 9B) . In particular, Helicobacter ganmani, Helicobacter hepaticus and Candidatus Borkfalkia ceftriaxoniphila were also inversely correlated to tumor count < 2mm (p < 0.05) , and Desulfovibrio porci and Candidatus Borkfalkia ceftriaxoniphila were inversely correlated to tumor count ≥ 2mm (p < 0.05) (Fig. 9B) . Noteworthily, Prohep elevated Helicobacter typhlonius inversely correlated to tumor count < 2mm (p < 0.05) , and proinflammatory cytokines TNF-α, IL-17A/F and IL-6 (p < 0.05) while being positively correlated to the abundance of fecal butyrate, propionate and acetate (p < 0.05) (Fig. 9B) . A correlation analysis between the enriched species was carried out to study the possible role of bacteria among the microbiota community of AOM/DSS and Prohep. Among the enriched species, Desulfovibrio porci, Prevotella sp. PTAC, Helicobacter ganmani and Muribaculaceae bacterium Isolate-007 (NCI) were found to be negatively correlated to AOM/DSS enriched bacteria inducing Clostridum sp. CAG: 510, bacterium 0.1xD8-71, Candidatus Gastranaerophilus sp. (ex Termes propinquus) , Vermiculatibacterium agrestimuris and Roseburia sp. CAG: 309 (p < 0.05) (Fig. 10) . Among them, Prevotella sp. PTAC demonstrated the highest number of correlations with other bacteria and negatively correlated with most of the AOM/DSS enriched bacteria and positively correlated with other Prohep enriched bacteria. In particular, Prevotella sp. PTAC and Desulfovibrio porci were significantly correlated with each other (p < 0.05) (Fig. 10) . The enriched Desulfovibrio porci was the only species found to be negatively correlated with all AOM/DSS enriched species and total tumor count at the same, highlighting its importance in attenuating CRC gut dysbiosis and tumorigenesis.
Furthermore, the possible functions of Prohep on governing metabolic and biosynthesis pathways in comparison with AOM/DSS were studied from Metacyc and KEGG analysis. For the Metacyc analysis, Prohep downregulated 17 pathways and upregulated 26 pathways (Fig. 11A) . In particular, Prohep reduced peptidoglycan biosynthesis II (staphylococci) (p < 0.05) and dTDP-3-acetamido-α-D-fucos biosynthesis (p < 0.05) , which were related to the biosynthesis of the detrimental peptidoglycan and lipopolysaccharide (LPS) . Prohep also downregulated purine nucleotides degradation II (aerobic) (p < 0.05) , which limits the conversion of purine into cancer related uric acid. In addition, Prohep elevated pathways related to the biosynthesis of beneficial compounds, including L-lysine biosynthesis I (p < 0.05) and L-lysine biosynthesis II (p < 0.05) which were related to the biosynthesis of L-lysine; octanoyl- [acyl-carrier protein] biosynthesis (mitochondria, yeast) (p < 0.05) for the biosynthesis of lipoic acid; pyrimidine deoxyribonucleotides biosynthesis from CTP (p < 0.05) , superpathway of pyrimidine deoxyribonucleotides de novo biosynthesis (p < 0.05) and superpathway of pyrimidine ribonucleotides de novo biosynthesis (p < 0.05) for the biosynthesis of pyrimidine; Rubisco shunt (p < 0.05) and 2-methylcitrate cycle I (p <0.05) for SCFA precursor pyruvate; and palmitate biosynthesis II (bacteria and plants) (p <0.05) , superpathway of unsaturated fatty acids biosynthesis (E. coli) (p < 0.05) and palmitate biosynthesis I (animals and fungi) (p < 0.05) which related to palmitate biosynthesis. Furthermore, Prohep also upregulated pathways energy utilization pathway of lactic-acid producing bacteria, lactose and galactose degradation I (p < 0.05) and TCA cycle VII (acetate-producers) (p < 0.05) which indicates for greater abundance of acetic acid producing bacteria.
In terms of the KEGG analysis, Prohep downregulated 23 KO gene and upregulated 55KO gene (Fig. 11B) . Especially, Prohep decreased arylformamidase (p < 0.05) , phosphoserine aminotransferase (p < 0.05) , beta-glucuronidase (p < 0.05) , alcohol dehydrogenase (NADP+) (p < 0.05) , transketolase (p < 0.05) and purine-nucleoside phosphorylase (p < 0.05) . In addition, Prohep increased aminodeoxyfutalosine deaminase (p < 0.05) , aminodeoxyfutalosine synthase (p < 0.05) , 1, 4-dihydroxy-6-naphthoate synthase (p <0.05) , chorismate dehydratase (p < 0.05) , cyclic dehypoxanthinyl futalosine synthase (p < 0.05) , adenosylhomocysteine/aminodeoxyfutalosine nucleosidase (p < 0.05) , acetyl-CoA acyltransferase (p < 0.05) , hydroxymethylbilane synthase (p < 0.05) , porphobilinogen synthase (p < 0.05) , glutamate-1-semialdehyde 2, 1-aminomutase (p < 0.05) , glutamyl-tRNA reductase (p < 0.05) , fructose-1, 6-bisphosphatase I (p < 0.05) , fructose-1, 6-bisphosphatase II (p < 0.05) , and 2-amino-4-hfigureydroxy-6-hydroxymethyldihydropteridine diphosphokinase /dihydropteroate synthase (p < 0.05) .
The fecal SCFA concentration was accessed based on the results of Metacyc analysis. In comparison with the healthy group, the fecal acetate concentration was lowered in the AOM/DSS, 5-FU and Prohep +5-FU groups (p < 0.05) (Fig. 9C) . Meanwhile, the fecal acetate concentration in Prohep was significantly increased compared to the AOM/DSS group (p < 0.05) . Furthermore, the relative abundance of acetate-producing Acidiphilium genus and Acidiphilium sp. CAG: 727 were upregulated by Prohep in comparison to AOM/DSS (p < 0.05) .
Discussion
In this study, Prohep significantly alleviated AOM/DSS-induced colorectal tumorigenesis by suppressing proliferative STAT3, inducing apoptotic p53, modulating the gut microbiota, and elevating acetate concentration. Prohep also demonstrated the strongest alleviative effects compared to 5-FU, and Prohep+5-FU. In the current study, all experimental mice developed colorectal tumors from the AOM/DSS treatment. Meanwhile, some mice in AOM/DSS, 5-FU, and Prohep+5-FU groups reached humane endpoints that required euthanasia as determined by animal facility veterinarians. Notably, only Prohep significantly improved the survivability of experimental animals, but not in 5-FU and Prohep+5-FU groups. Although all three treatment groups, including 5-FU, Prohep and Prohep+5-FU, significantly reduced the total tumor count, only Prohep significantly reduced the total tumor size as well. 5-FU was the only group that was not able to reduce the enlarged caecum. In addition, 5-FU and Prohep+5-FU led to a reduction in liver weight in comparison to the healthy group. As the liver is the major organ for metabolizing 5-FU, the administration of 5-FU was suggested to cause liver function failure and hepatotoxicity including apoptosis, inflammation, and collagen fibrosis, as seen with the reduced liver weight. Prohep was the only group that significantly improved survivability and reduced tumor count, size and caecum weight when compared to the AOM/DSS group. Therefore, it was suggested that Prohep was an effective therapeutic alternative as it protected liver functions while also being anti-CRC.
In this study, Prohep demonstrated anti-inflammatory responses in the colon. Inflammation-related histopathological parameters, including hyperplasia, disrupted colonic structure, lengthened colonic crypt depth, and collagen fibrosis, were all alleviated by Prohep. In addition, the pro-inflammatory cytokines TNF-α and inflammatory regulator STAT3 activation were suppressed. Besides the well-known proinflammatory IL-6 cytokine, TNF-αwas found to promote proliferation and inhibit apoptosis in colon cancer cells by activating STAT3. STAT3 was suggested to be a mediator connecting inflammation and cancer progression, in which silencing of the STAT3 was found to limit CRC cell growth and induce cell death at the G2/M stage. Prohep also downregulated c-jun and c-fos expression levels which functioned as the effectors of STAT3-induced tumor cell proliferation. Furthermore, STAT3 silencing also led to apoptosis through upregulation of p53 and caspase-3. Similar results were observed in the present study with the elevated expression level of p53 and c-casp3. p53 serves an important function in suppressing tumor growth by inducing cell cycle arrest, DNA repair, senescence and apoptosis. Deficiencies of p53 were often found in cancer and the restoration of p53 was targeted for cancer therapy including CRC. At the same time, p53 also inhibits STAT3 activators and attenuates STAT3 functions. Given both STAT3 and p53 are effective targets and the presence of opposing STAT3-p53 regulatory loops, co-targeting STAT3 and p53 by Prohep was therefore suggested to be promising in CRC regulation.
Apart from its effects on cell signaling pathways, Prohep administration also altered the gut microbiota. Prohep enriched the relative abundance of Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila and Helicobacter typhlonius, which showed a reverse correlation with colorectal tumor counts in this study. Helicobacter ganmani and Helicobacter hepaticus are two Helicobacter species naturally colonizing mouse gut which are reported to be related to IBD development. Nevertheless, a recent study suggested that Helicobacter ganmani and the augment of Helicobacter enhanced the induction of RORγt+ Foxp3+ iTregs cells which improved the resistance against DSS-induced colitis in a fecal microbiota transplant (FMT) model pretreated with glycerol monolaurate. Further studies on the potential beneficial effects of Helicobacter ganmani in the colon are necessary to reconcile the contradicting observations. Similarly, a previous study described that Helicobacter typhlonius when co-administrated with Akkermansia muciniphila would reduce intestinal tumors in Apc mutant mice while opposing results were observed when either bacterium was singly administrated. Moreover, Desulfovibrio porci was also found to be inversely correlated to tumor count. Except for identifying it as a hydrogen sulfide (H2S) producing species, its effects on animals were not well-documented. H2S was reported with dual effects on cancer cells following a biphasic dose-response curve. Recent studies suggest that H2S exposure in high concentrations for prolonged periods induced apoptotic responses in cancer cells both in vivo and in vitro. Thus, it was proposed that the H2S-producing properties of Desulfovibrio porci might contribute to the alleviation of colorectal tumorigenesis in this study which is worth further validation in the future. Furthermore, the enriched Candidatus Borkfalkia ceftriaxoniphila, a newly discovered and low abundant commensal species, had a significant increase in abundancies accompanying the growth of probiotics in the human gut.
The inner correlation analysis identified Prohep-enriched Desulfovibrio porci, Prevotella sp. PTAC, Helicobacter ganmani and Muribaculaceae bacterium Isolate-007 (NCI) were inversely correlated to AOM/DSS enriched bacteria. Both Desulfovibrio porci and Prevotella sp. PTAC displayed a significant role in Prohep-induced gut microbiota modulation and a positive correlation in relative abundance was found between them. A higher abundance of Prevotella was reported to be linked to lower risks of CRC progression in CRC patients. Although Prevotella sp. PTAC was not found to be significantly correlated to reduced tumor count in this study, its co-occurrence with Desulfovibrio porci and other Prohep-enriched bacteria suggested it played a vast role in supporting the growth of these beneficial bacteria in CRC management. Notably, only the enrichment of Desulfovibrio porci negatively correlated to all AOM/DSS enriched bacteria. Given its negative correlation with CRC tumor count as well, the upregulation of Desulfovibrio porci was suggested to be crucial in alleviating CRC tumorigenesis through reversing AOM/DSS-induced gut microbiota.
Apart from the correlation against the colorectal tumorigenesis-related parameters, the mechanisms behind the anti-CRC effects of Prohep was also studied through the prediction of the metabolic functions from Metacyc and KEGG analysis. From the Metacyc analysis, Prohep downregulated the CRC-related peptidoglycan and LPS biosynthesis as well as the conversion of uric acid from purine. Peptidoglycan is a major component of the gram-positive bacteria cell wall. When peptidoglycan is transferred across the intestinal epithelial cells, it interacts with macrophages, triggers IL-6 secretion and induces inflammation and fibrosis in Crohn’s disease and CRC. Similarly, LPS is a cellular component from bacterial membranes which is known for its endotoxic properties. LPS would induce inflammation by activating nuclear factor-κB (NF-κB) pathway via Toll-like receptor 4 (TLR4) which exacerbates gut barrier dysfunction and promotes CRC development. In addition, in the catabolism of purine nucleotides, uric acid is released which exhibits a positive association with CRC incidence .
Prohep, on the other hand, elevated the biosynthesis pathways of beneficial compounds including L-lysine, lipoic acid, pyrimidine, and palmitate. While L-lysine does not contribute directly to cancer management, it could be completely be converted into butyrate and acetate by gut microbiota like Intestinimonas strain AF211 and served a significant role in inhibiting CRC and maintaining the growth of normal colonocytes. Meanwhile, the combination of lysine with epigallocatechin gallate (EGCG) were found to enhance the inhibitory effects of EGCG on the growth of colon cancer cell line HCT 116. Alpha-lipoic acid (LA) was found to have anti-CRC by triggering cell death in tumor cells and could be combined with doxorubicin or 5-FU to synergistically kill CRC cells. LA also prevent inflammatory and oxidative responses by suppressing the NF-κB pathway, as well as downregulating TNF-α, IL-6, COX-2, MDA, and MPO. Pyrimidine and its derivatives were reported with pharmacological and anti-cancer properties, in which pyrimidine metabolic pathways were reported to regulate the chemotherapeutic effects of chemo drugs like 5-FU, tegafur and thioguanine. In particular, the derivative N- [2- (dimethylamino) ethyl] -2, 3-dimethyl-4-oxo-4H-pyrido [1, 2-a] thieno [2, 3-d] pyrimidine-9-carboxamide (PTP) showed strong antitumor effects against human CRC cells by activating p53. Palmitate is one of the common saturated fatty acids found in animals and plants which previous studies reported that a low intake of palmitic acid showed inverse associations with CRC and palmitic acid alongside ceramide were strong inhibitors of the EMT signaling axis of colorectal cancer cells. The upregulation of Rubisco shunt and 2-methylcitrate cycle II both supported the production of pyruvate which is a precursor of acetate and butyrate. Through pyruvate decarboxylation, pyruvate is oxidated to the intermediate acetyl-CoA, which could produce acetate via acetate kinase or condense with another acetyl-CoA and be further reduced for butyrate.
Moreover, Prohep elevated metabolic pathways related to energy utilization of lactic acid-producing bacteria (LAB) and acetate producers. Since Prohep is composed mostly of LAB, the increase of lactose and galactose degradation was expected and intestinal galactose was reported to have a protective effect against CRC by inhibiting mucosal proliferation. Besides, the TCA cycle of acetic acid bacteria was enhanced by Prohep. Acetic acid bacteria are known for their properties in oxidizing ethanol to acetic acid and the abundance of acetate-producing Acidiphilium was elevated. Acetate was identified as one of the important metabolites which inhibited CRC tumorigenesis in both in vivo and in vitro models. Acetate reduced the tumor size of a CRC-cell-injected xenograft mouse model. It was suggested that acetate enhanced growth arrest and apoptosis of CRC cells through increasing oxygen consumption and reactive oxygen species production. Previous studies demonstrated more pronounced apoptotic effects on CRC cells compared to normal colonocytes, achieved by enhancing MCT1, MCT4 and CD147 while also re-localizing MCT1 at the plasma membrane. Also, acetate would serve as an energy substrate for normal colonocytes via de novo lipogenesis and allows the normal cells to outcompete cancer cells, which primarily rely on glycolysis for energy production. Given the elevation of pathways related to SCFAs and acetate production from Metacyc analysis, the fecal SCFAs profile was evaluated. Prohep significantly enriched acetate concentration in this study. Similar to the previous study, Prohep increased fecal acetate level to achieve hepatic lipid regulation in the MASLD/MASH model. The results disclosed herein results suggest that acetate might play a role in the overall reduction of colorectal tumorigenesis.
In terms of the KEGG analysis, Prohep downregulated the detrimental arylformamidase, phosphoserine aminotransferase, beta-glucuronidase, alcohol dehydrogenase (NADP+) , transketolase and purine-nucleoside phosphorylase pathways. Arylformamidase (AFMID) was involved in the conversion of tryptophan into kynurenine by the transcription factor MYC in both cultured CRC cells and CRC patients. Phosphoserine aminotransferase is responsible for serine biosynthesis and an elevated serine was suggested to enhance the growth of CRC cells. The overexpression of phosphoserine aminotransferase was reported to heighten CRC cells chemoresistance and cell growth. The serum activity of beta-glucuronidase was found to be higher in CRC patients than in healthy subjects, which suggested it as a marker of CRC. In the ascorbate-dependent alcohol oxidation system, alcohol dehydrogenase (NADP+) facilitated oxygen to be consumed and hydrogen peroxide (H2O2) to form. The level of H2O2 in the tumor microenvironment was closely associated with the development of CRC. Transketolase was abnormally increased in CRC which promoted cancer cell glycolysis by enhancing AKT phosphorylation and eventually worsening CRC metastasis. Purine-nucleoside phosphorylase (PNP) was identified as a cancer marker as plasma PNP levels on average four times higher in cancer patients. For CRC, the expression level of PNP was also correlated with lymph vessel invasion, positive lymph node metastasis and advanced stage of CRC. The downregulation of these CRC-related pathways strengthened Prohep’s promising effects in inhibiting colorectal tumorigenesis.
Moreover, Prohep elevated multiple beneficial pathways in the regulation of CRC. Aminodeoxyfutalosine deaminase, aminodeoxyfutalosine synthase, 1, 4-dihydroxy-6-naphthoate synthase, chorismate dehydratase, cyclic dehypoxanthinyl futalosine synthase, and adenosylhomocysteine/aminodeoxyfutalosine nucleosidase are related to menaquinones which are the bacterial forms vitamin K produced by gut microbiota. Menaquinone or vitamin K2 was found to reduce KRAS proliferation in colon cancer cells and promote apoptotic cell death in CRC mice. Acetyl-CoA acyltransferase (ACAA) was shown to have a negative correlation with the resistance of the targeted cancer drug, cetuximab, in CRC which the overexpression of ACAA would suppress proliferation and lower cetuximab tolerance in CRC cells. Hydroxymethylbilane synthase was identified as a tumor suppressor gene and its inactivation was identified in patients with intermittent porphyria and sporadic HCC. Porphobilinogen synthase is the crucial first step of tetrapyrrole biosynthesis which tetrapyrroles like unconjugated bilirubin, bilirubin ditaurate, biliverdin, biliverdin-/bilirubin dimethyl ester, urobilin, stercobilin and protoporphyrin exhibited DNA-damaging and apoptosis in colon and liver cancer cells. Meanwhile, porphobilinogen synthase, along with glutamate-1-semialdehyde 2, 1-aminomutase and glutamyl-tRNA reductase are involved in the production and utilization of aminolevulinic acid (ALA) which was shown to inhibit CRC cells. Fructose-1, 6-bisphosphatase I could inactive NF-κB which suppresses CRC and similarly, fructose-1, 6-bisphosphatase II was found to regulate gastric cancer in an inversed relationship. 2-amino-4-hydroxy-6 hydroxymethyldihydropteridine diphosphokinase/dihydropteroate synthase was required in the biosynthesis of tetrahydrofolate which was associated with lowered risk of serrated polyps in CRC. These results suggested that the gut microbiota modulation of Prohep was important in establishing an anti-CRC metabolic profile.
In current study, Prohep was found to be the most effective treatment for AOM/DSS mice when compared with 5-FU and Prohep+5-FU. No additive or synergistic effects were observed when Prohep was given in adjuvant with 5-FU. Prohep presented the strongest inhibition on colorectal tumorigenesis and inflammation among all groups. In the present study, even though 5-FU reduced the total tumor count in the colon, it failed to reduce the level of p-STAT3 and upregulate that of p53. The activation of STAT3 was reported to promote 5-FU resistance in CRC through increasing Mcl-1-dependent cytoprotective autophagy, by which 5-FU resistant cells would transfer p-STAT3-containing exosomes to the recipient cells and induce chemoresistance against 5-FU. On the other hand, the loss or deficiency of p53 would contribute to 5-FU resistance and detriment the DNA damaging effects of 5-FU against CRC cells. The restoration of p53 was reported to significantly improve 5-FU sensitivity in CRC cells. Nonetheless, even though Prohep increased p53 and reduced p-STAT3, the effects were not displayed when Prohep was co-treated with 5-FU. Besides a disrupted gut microbiome environment, 5-FU could lead to gut dysbiosis and intestinal mucositis. These effects may hinder the adhesion, colonization and activation of probiotic species and might thus limit the anti-CRC effects of Prohep when co-administrated.
Conclusion
In conclusion, the findings demonstrated that Prohep reduced AOM/DSS-induced CRC carcinogenesis. Through suppressing STAT3, and activating apoptotic-inducing p53, Prohep significantly reduced total tumor count, total tumor size, caecum weight, colonic crypt depth, colonic inflammation, and collagen fibrosis induced by AOM/DSS. Furthermore, Prohep enriched the abundance of beneficial bacteria and acetate level which contributed to combat CRC. Notably, Prohep showed superior anti-tumorigenesis effects compared to both 5-FU alone and Prohep+5-FU in the treatment of CRC. These findings indicate Prohep to be an effective alternative treatment for CRC.
Reference
Leung, H. K. M., Lo, E. K. K., Chen, C., Zhang, F., Ismaiah, M. J., &El-Nezami, H. (2024) . Probiotic Mixture Attenuates Colorectal Tumorigenesis in Murine AOM/DSS Model by Suppressing STAT3, Inducing Apoptotic p53 and Modulating Gut Microbiota. Probiotics and Antimicrobial Proteins, 1-17.
Abbreviations

It is understood that the disclosed method and compositions are not limited to the particular methodology, protocols, and reagents described as these can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the method and compositions described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (22)

  1. A composition comprising a probiotic formulation, wherein the probiotic formulation comprises one or more microbiota selected from a group consisting of Lactobacillus helveticus, Lactobacillus plantarum, Lactobacillus rhamnosus GG, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium animalis subsp. Lactis, Streptococcus thermophilus or any combination thereof.
  2. The composition of claim 1 further comprising an anti-cancer agent.
  3. The composition of claim 2, wherein the anti-cancer agent comprises one or more of chemotherapeutics, targeted therapies, immunotherapies, hormonal therapies, epigenetic modulators, radiopharmaceuticals, angiogenesis inhibitors, natural products, nanoparticle-based therapies, RNA-based therapies, microbiome-modulating treatments, or any combination thereof.
  4. The composition of claims 2 or 3, wherein the anti-cancer agent is a sorafenib.
  5. The composition of claim 4, wherein the sorafenib is in a dose in a range of about 30 mg/kg –800 mg/kg body weight, 25 mg/kg –700 mg/kg body weight, 20 mg/kg –600 mg/kg body weight, 15 mg/kg –500 mg/kg body weight, 10 mg/kg –400 mg/kg body weight, 5 mg/kg –300 mg/kg body weight, 1 mg/kg –200 mg/kg body weight.
  6. The composition of claim 4 or 5, wherein the dose of sorafenib is about 30 mg/kg body weight.
  7. A method of treating a subject, the method comprising administering to the subject an amount of the composition of any one of claims 1-6 effective to:
    (a)
    (i) treat or prevent metabolic dysfunction-associated steatotic liver disease-associated hepatocellular carcinoma (MASLD-HCC) ,
    (ii) modulate gut microbiota of the subject to increase production of short-chain fatty acids (SCFAs) ,
    (iii) activate AMPK signaling resulting in reduced lipogenesis, decreased lipid uptake, and upregulation of antioxidant enzyme expressions,
    (iv) suppress the PI3K/mTOR cancer proliferation pathway,
    (v) reduce tumor counts, ameliorate inflammation, and/or increase hepatic superoxide dismutase (SOD) expression, or
    (vi) a combination thereof; or
    (b)
    (i) treat or prevent colorectal cancer (CRC) ,
    (ii) alleviate AOM/DSS-induced colorectal tumorigenesis,
    (iii) modulate inflammatory, proliferative, and/or apoptotic pathways,
    (iv) downregulate TNF-α and p-STAT3 and/or upregulates p53,
    (v) reduce tumor burden, caecum weight, crypt depth, colonic inflammation, and/or collagen fibrosis induced by AOM/DSS,
    (vi) enrich beneficial bacteria such as Helicobacter ganmani, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila, Desulfovibrio porci, and Prevotella sp. PTAC in the subject,
    (vii) upregulate fecal acetate concentration, or
    (viii) a combination thereof.
  8. The method of claim 7, wherein the subject has MASLD-HCC or has been identified as being at risk of developing MASLD-HCC.
  9. The method of claim 7 or 8, wherein the composition comprises the anti-cancer agent, wherein the composition is administered in an amount effective to treat or prevent MASLD-HCC.
  10. The method of any one of claims 7-9, wherein the composition modulates gut microbiota of the subject to increase production of short-chain fatty acids (SCFAs) .
  11. The method of any one of claims 7-10, wherein the composition activates AMPK signaling resulting in reduced lipogenesis, decreased lipid uptake, and upregulation of antioxidant enzyme expressions.
  12. The method of any one of claims 7-11, wherein the composition suppresses the PI3K/mTOR cancer proliferation pathway, contributing to reduced tumor counts, amelioration of inflammation, and increased hepatic superoxide dismutase (SOD) expression.
  13. The method of any one of claims 7-12, wherein the composition comprises sorafenib, wherein the composition improves therapeutic outcomes in treating MASLD-HCC compared to treatment with sorafenib alone.
  14. The method of any one of claims 7-13, wherein the composition modulates gut microbiota to increase sensitivity of MASLD-HCC to sorafenib.
  15. The method of claim 7, wherein the subject has CRC or has been identified as being at risk of developing CRC.
  16. The method of claim 7 or 15, wherein the composition treats or prevents CRC.
  17. The method of claim 7, 15, or 16, wherein the composition alleviates AOM/DSS-induced colorectal tumorigenesis by modulating inflammatory, proliferative, and/or apoptotic pathways, preferably by downregulation of TNF-α and p-STAT3 and/or upregulation of p53.
  18. The method of any one of claims 7 or 15-17, wherein the composition reduces tumor burden, caecum weight, crypt depth, colonic inflammation, and/or collagen fibrosis induced by AOM/DSS.
  19. The method of any one of claims 7 or 15-18, wherein the composition enriches beneficial bacteria such as Helicobacter ganmani, Helicobacter hepaticus, Candidatus Borkfalkia ceftriaxoniphila, Desulfovibrio porci, and Prevotella sp. PTAC in the subject.
  20. The method of any one of claims 7 or 15-19, wherein the composition upregulates fecal acetate concentration.
  21. The method of any one of claims 7 or 15-20, wherein the composition enhances anti-cancer effects compared to 5-fluorouracil (5-FU) alone or to the combination of the composition and 5-FU in the treatment of CRC.
  22. The method of any one of claims 7-21, wherein the composition is administered orally, intracolonically, intranasally, intrarectally, via a catheter, via a lavage, via a nasogastric tube, via local delivery, via a method for fecal microbiota transplantation (FMT) , or any combination thereof.
PCT/CN2025/078098 2024-02-19 2025-02-19 Probiotics mixtures and methods of use thereof for treating cancers Pending WO2025176152A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202463555341P 2024-02-19 2024-02-19
US202463555348P 2024-02-19 2024-02-19
US63/555,341 2024-02-19
US63/555,348 2024-02-19

Publications (1)

Publication Number Publication Date
WO2025176152A1 true WO2025176152A1 (en) 2025-08-28

Family

ID=96846475

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2025/078098 Pending WO2025176152A1 (en) 2024-02-19 2025-02-19 Probiotics mixtures and methods of use thereof for treating cancers

Country Status (1)

Country Link
WO (1) WO2025176152A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105658226A (en) * 2013-08-16 2016-06-08 香港大学 Methods and compositions for treating cancer using probiotics
CN107405321A (en) * 2015-01-23 2017-11-28 坦普尔大学 Application of short-chain fatty acids in cancer prevention
CN107847530A (en) * 2015-06-01 2018-03-27 芝加哥大学 By controlling symbiotic microorganism clump come treating cancer
WO2020041581A1 (en) * 2018-08-23 2020-02-27 Cornell University Methods and compositions for preventing and treating inflammatory bowel disease and nonalcoholic fatty liver disease
CN114806979A (en) * 2022-06-29 2022-07-29 杭州师范大学附属医院(杭州市第二人民医院) Probiotic compound for treating non-alcoholic fatty liver disease and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105658226A (en) * 2013-08-16 2016-06-08 香港大学 Methods and compositions for treating cancer using probiotics
CN107405321A (en) * 2015-01-23 2017-11-28 坦普尔大学 Application of short-chain fatty acids in cancer prevention
CN107847530A (en) * 2015-06-01 2018-03-27 芝加哥大学 By controlling symbiotic microorganism clump come treating cancer
WO2020041581A1 (en) * 2018-08-23 2020-02-27 Cornell University Methods and compositions for preventing and treating inflammatory bowel disease and nonalcoholic fatty liver disease
CN114806979A (en) * 2022-06-29 2022-07-29 杭州师范大学附属医院(杭州市第二人民医院) Probiotic compound for treating non-alcoholic fatty liver disease and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JUN LI ETC.: "Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice", PROCEEDINGS OF THE NATIONAL ACACEMY OF SCIENCE OF THE UNITED STATES OF AMERICA, vol. 113, no. 9, 1 March 2016 (2016-03-01), XP055454032, DOI: 10.1073/pnas.1518189113 *
ZHANG FANGFEI, LO EMILY KWUN KWAN, CHEN JIARUI, WANG KE, FELICIANNA, ISMAIAH MARSENA JASIEL, LEUNG HOI KIT MATTHEW, ZHAO DANYUE, L: "Probiotic Mixture Ameliorates a Diet-Induced MASLD/MASH Murine Model through the Regulation of Hepatic Lipid Metabolism and the Gut Microbiome", JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 72, no. 15, 17 April 2024 (2024-04-17), US , pages 8536 - 8549, XP093347728, ISSN: 0021-8561, DOI: 10.1021/acs.jafc.3c08910 *

Similar Documents

Publication Publication Date Title
Liu et al. Phellinus linteus polysaccharide extract improves insulin resistance by regulating gut microbiota composition
Ferrere et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade
Ghosh et al. Microbial metabolite restricts 5-fluorouracil-resistant colonic tumor progression by sensitizing drug transporters via regulation of FOXO3-FOXM1 axis
Villanueva-Millán et al. Gut microbiota: a key player in health and disease. A review focused on obesity
JP2023010890A (en) Methods for treating ulcerative colitis
Ji et al. The gut microbiome and ferroptosis in MAFLD
Zhu et al. Riboflavin bioenriched soymilk alleviates oxidative stress mediated liver injury, intestinal inflammation, and gut microbiota modification in B2 depletion–repletion mice
Zhao et al. Protective effects of Lactobacillus plantarum C88 on chronic ethanol-induced liver injury in mice
Cavalcanti et al. Administration of reconstituted polyphenol oil bodies efficiently suppresses dendritic cell inflammatory pathways and acute intestinal inflammation
Feng et al. Pentamethoxyflavanone regulates macrophage polarization and ameliorates sepsis in mice
Wang et al. Ginkgo biloba extract ameliorates atherosclerosis via rebalancing gut flora and microbial metabolism
Sreenivasan Snima et al. Anti-diabetic drug metformin: challenges and perspectives for cancer therapy
Pan et al. Role of gut microbiota in the pharmacological effects of natural products
Wang et al. Biomimetic nanoplatform with microbiome modulation and antioxidant functions ameliorating insulin resistance and pancreatic β-cell dysfunction for T2DM management
Gu et al. Role of gut microbiota and bacterial metabolites in mucins of colorectal cancer
Araki et al. Genipin attenuates lipopolysaccharide-induced persistent changes of emotional behaviors and neural activation in the hypothalamic paraventricular nucleus and the central amygdala nucleus
WO2018078539A1 (en) Pharmaceutical composition for use in the therapeutic treatment of cancer and complications of cancer
JP2024540177A (en) Methods of Treating Endometrial Cancer Using Hemp Extract
Ai et al. Exploring the phosphatidylcholine in inflammatory bowel disease: potential mechanisms and therapeutic interventions
Zhang et al. A steamed broccoli sprout diet preparation that reduces colitis via the gut microbiota
Kapoor et al. Phloretin suppresses intestinal inflammation and maintained epithelial tight junction integrity by modulating cytokines secretion in in vitro model of gut inflammation
Zhang et al. The biological activity and potential of probiotics-derived extracellular vesicles as postbiotics in modulating microbiota-host communication
Zhu et al. Inulin‐based nanoparticle modulates gut microbiota and immune microenvironment for improving colorectal cancer therapy
Yao et al. Exosomes: Mediators in microenvironment of colorectal cancer
Meng et al. Chemopreventive effect of the polysaccharides from Grifola frondosa in colitis-associated colorectal cancer by modulating the Wnt/β-catenin/GSK-3β signaling pathway in C57BL/6 mice

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 25757744

Country of ref document: EP

Kind code of ref document: A1