COMPOSITIONS OF AZADIRACHTA INDICA AND METHODS OF TREATING CANCER

Abstract
The disclosure relates to compositions and methods of treating cancer in a subject. The method comprises administering to a patient in need of treatment an effective amount of supercritical CO2 neem extract. The disclosure also relates to a process for preparing a CO2 extract of Azadirachta indica and herbal compositions thereof for the treatment of oral and colon cancers. The methods comprise a process for preparing a standardized SCO2 extract of Azadirachta indica leaves and herbal compositions of the same for oral use.
Description
FIELD

The present disclosure relates to process for preparation of CO2 extract of Azadirachta indica and herbal compositions thereof for the treatment of oral and colon cancers. More particularly, the disclosure relates to a process for preparation of standardized SCO2 extract of Azadirachta indica leaves and herbal compositions of the same for oral use.


BACKGROUND

The risk for developing oral cancers is increasing world-wide as the global rise in tobacco use, alcohol consumption, and HPV exposure continues. Conventional treatments have improved the 5-year survival rates for patients with early disease, while patients with late-stage disease have a 5-year survival rate as low as 34% which has not changed in nearly 40 years.


Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second leading cause of cancer death in men and women in the United States. Anti-inflammatory blockade has been proven to be a promising avenue of colorectal cancer prevention. However, NSAIDs, while effective in curbing CRC risk, are too toxic for long term use in cancer prevention.


Oral cancers are among the top three types of cancers in India. It appears as a growth or sore in the mouth that does not cure and includes cancers of the lips, tongue, cheeks, floor of the mouth, hard and soft palate, tongue, sinuses, and pharynx. The most common type of oral cancer is Squamous cell carcinoma.


According to the statistics, in 2012 the incidence of oral cancer in India was 53842 in males and 23161 in females. In India, the prevalence is high (20/100,000 population) and incidence is expected to rise by 2030. The international agency for research on cancer has predicted that India's incidence of cancer is from 1 million in 2012 and likely to be more than 1.7 million in 2035. This indicates that the death rate because of cancer will also increase from 1 million to 1-2 million in the same period. Incidence of oral cancer is more in men. Its incidence also increases by age with most of the oral cancer occurring in 50 to 70 years of age.


Globally, lip, oral cavity, and pharyngeal cancers had accounted for about 3.8% of all cancer cases and 3.6% of overall cancer deaths as per 2012 data. According to GLOBOCAN 2012, lip and oral cavity cancer is the 12th most common cancer in Asia and ranks 8th among cancers in men. In Asia and America, the incidence rates are 3.8% and 1.7% respectively, while mortality rate are 2.2% and 1% respectively.


Various etiological factors like tobacco consumption either smokeless tobacco or smoking and alcohol consumption had attributed to the high incidence of oral cancer in India. Also, positive family history of oral cancer, viral infections like HPV, poor oral hygiene, dietary deficiencies, and oro-dental factors are the other contributing factors.


Oral cancer can be detected by symptoms like pain in throat, long-standing ulcers in the mouth, loosening of teeth, and change in voice and difficulty in chewing and swallowing.


Colorectal cancer (CRC), also known as colon cancer or bowel cancer, is one of the leading causes of mortality and morbidity from cancer. Worldwide, it is the third most common cancer in men (10.0% of cancer cases) and the second most common in women (9.4% of cancer cases) with 60% of the cases encountered in developed countries. Every year, there are 1.3 million new cases of CRC globally with a 5-year prevalence rate of 3.2 million. In India, though the incidence of CRC has increased marginally, it is now the fifth most common cause of cancer mortality among Indian men and women. In India, the annual incidence rates for colon cancer and rectal cancer in men are 4.4 and 4.1 per 100000, respectively and the annual incidence rate for colon cancer in women is 3.9 per 100000. Thus, colon cancer ranks 8th and rectal cancer ranks 9th among men while for women, rectal cancer does not figure in the top 10 cancers, whereas colon cancer ranks 9th.


Risk factors for CRC include age (risk of colorectal cancer increases with age), gender (25% higher in men than in women), personal history of colorectal polyposis, inflammatory bowel disease (ulcerative colitis or Crohn's disease), family history of colorectal cancer and lifestyle related factors like obesity, alcohol consumption and cigarette smoking. Being physically inactive also increases the risk of developing colorectal cancer.


Signs of colorectal cancer are change in bowel habit, sensation of incomplete emptying after passing motion, blood mixed with stool, passing mucus with stool, sensation of fullness after eating is less, abdominal distension, abdominal pain, weight loss, constipation, diarrhea, frequent urge to pass stool, fatigue, vomiting, bloating and pain in the abdomen, iron deficiency and a lump in the stomach.


Surgery, chemotherapy, and radiotherapy are still the major conventional cancer therapies. However, more than 50% of patients have minimal or no benefit from these treatments and most of them suffer from their toxic adverse reactions. Alternative medicine (like herbal medicines) has become increasingly popular among cancer patients with a prevalence of its use as high as 80%.


Neem also known as Azadirachta indica is commonly found in many semi-tropical and tropical countries including India. The components extracted from neem plant have been used in traditional medicine for the cure of multiple diseases including cancer for centuries. Studies have shown compelling evidence suggesting that the anticancer effects of neem are mediated through modulation of multiple cellular processes. Nimbolide, an active molecule isolated from Azadirachta indica, has been reported to exhibit several medicinal properties. It has shown potent anticancer activity against several types of cancer and has demonstrated potential anti-cancer activity in several in-vivo and in-vitro studies. Nimbolide acts by generating reactive oxygen species (ROS), thereby inducing apoptosis, inhibition of metastasis and angiogenesis. Another component nimbin, a triterpenoid isolated from Azadirachta indica possess anti-inflammatory, anti-pyretic, anti-histamine and anti-fungal properties.


WO2015035199A1 provides a method for treating one or more symptoms of cancer by administering a therapeutically effective dose of a pharmaceutical formulation to the patient to ameliorate one or more symptoms of the cancer or to reduce the number of cancer cells, wherein the pharmaceutical formulation comprises Nimbolide; Nimbandiol; 2′, 3′dihydro nimbolide; 28 dihydro nimbolide, or a combination thereof. The extract was examined on plasma and tumor tissues of mice at a dosage of 200 mg/kg of body weight administered orally. Targeted cancers were prostate cancer, colon cancer, astrocytoma and sarcoma. The invention provides bioactive compounds from Super Critical Neem Leaf Extract that exert anti-tumor activity. The patent claims the reduction of number of cancer cells, but the impact of reduction of cancer cells on symptoms of cancer is not known/determined/demonstrated. The extraction was carried out using supercritical CO2 at a pressure of 632.76 kg/cm2 and a temperature of 50° C. and collection was done at −49° C. using a dry ice/acetone bath. This huge difference between extraction temperature and separation temperature may cause unforeseen detrimental changes in the phytoconstituents. Also, the separation temperature is not commercially viable or scalable for industrial production.


CN101972246B provides an anti-tumor medicament which contains effective amount of Azadirachta indica triterpenoid 1 or 2 or 3 (see below) and a pharmaceutically acceptable carrier. It also provides a preparation method of a medicament which contains the compounds 1 to 3, and the application of the compounds to the preparation of medicaments for preventing and treating tumor diseases. It is provided to target leukemia, liver cancer, lung cancer and breast cancer. The extraction process was carried out by first subjecting herbs to methanol extraction and then subjecting the diluted methanolic extract to petroleum ether extraction and ethyl acetate extraction. The daily dosage varies from 0.01-10 mg/kg of body weight, with oral, parenteral, intrathecal or intraventricular administration.




embedded image


CN103864876A relates to new triterpenoid Xylocarpus granatum isolated from the neem tree fruit and use in the preparation of a medicine for malignant tumors particularly lung cancer and breast cancer. As stated in the patent application, pharmacological tests showed that the compound has inhibitory activity on the A-549 human lung adenocarcinoma cell line. Methanolic extract of neem tree foliage was used. It was also tested on human breast cancer cells A-549 for antitumor activity.


U.S. Pat. No. 5,370,873A relates to a purified extract of Azadirachta indica leaves which inhibit adhesion of cancer cells and malarial-infected erythrocytes to cultured endothelial cells. The purified extract also inhibits in-vitro vital development of human immunodeficiency virus (HIV), yellow fever virus and sandfly fever (Sicilian) virus and inhibits in vitro development of both sexual (gametocytes) and asexual (schizonts) forms of human malarial parasites. It also relates to a process for extracting purified extract from Neem leaves by soxhlet extraction, by a variety of solvents such as alcohols, acetone, pyridine, water etc. followed by passive precipitation and HPLC fractionation. The mechanism of working as stated is that due to anti-adhesive property of the neem leaf extract, the extract renders the cancer cells and infectious cells ineffective and non-proliferative.


JP2009274956A provides a composition which contains Epoxyazadiradione, gedunin, 17-epi-17-hydroxyazadiradione and 7-O-benzoylnimbocinol as active components in Neem seed extract to target leukemia, lymphomas, skin cancer, lung cancer, colon cancer, stomach cancer, breast cancer, prostate cancer, epithelial cell cancers such as thyroid cancer, bone sarcoma, etc. The extracts were obtained by using n-hexane followed by methanol and the fractionation by using column chromatography. The cytotoxic activity is based on apoptosis inducing activity of the compounds in the Neem seed extract. Also, it possesses anti-tumor activity. The test compounds were added to DMSO at concentrations of 1×10−4, 1×10−5, 1×10−6 M and its activity was tested. However, whether the effects are dose dependent, needs to be ascertained.


WO2007137389A1 provides a pharmaceutical composition for treatment of patients suffering with the human immunodeficiency virus. The composition can be administered orally. This composition comprises an effective amount of components obtained from Azadirachta indica; and a protein supplement. The composition is produced by immersing powder Neem in water to produce an aqueous extract.


Taken together, it was observed that, in conventional solvent extraction processes, the separation temperature is always greater than extraction temperature. The extraction of herb at higher temperature destroys or modifies molecular and chemical structure of temperature sensitive phytoconstituents which provide therapeutic value of herb. Also, it was seen from the art that the herb is extracted using solvents such as hexane, methanol, and acetone which are considered hazardous and carcinogenic for humans. Hence, there is a need for alternate but robust process for preparation of CO2 extract of Azadirachta indica in order to protect the heat sensitive phytoconstituents during extraction process and thereby therapeutic value of herbal formulation that is intended for treatment of cancers especially oral and colon cancers.


Thus, the objective of the present invention is to develop a robust process for obtaining Azadirachta indica leaf extract without compromising on the heat sensitive phytoconstituents.


SUMMARY

Disclosed herein are methods of treating cancer in a subject, the methods comprising: (a) identifying a subject in need of treatment; and (b) administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of reducing at least one inflammatory cytokine in serum of a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of reducing inflammation in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of treating a hyperproliferative disorder in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of suppressing expression of NFkB and cycloxygenase in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of suppressing expression of NFkB and cyclooxygenase in at least one cell, the methods comprising the step of contacting at least one cell with an effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of modifying epidermal growth factor receptor signaling activity in a subject, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


Disclosed herein are methods of inducing apoptosis of a cell in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.


The present disclosure provides a process for preparing Azadirachta indica leaf extract by using supercritical carbon dioxide (SCO2). The extract prepared can be administered either orally as capsules (veggie or hard gelatin capsules, liquid veggie capsules or soft gelatin capsules) or can be used as liquid mouthwash or sublingual oral formulation.


In a preferred aspect, the disclosure provides a process of preparing Azadirachta indica leaves extract containing beneficial phytoconstituents. The process involved following steps:

    • a) Clean and matured Azadirachta indica leaves are dried to reduce moisture to less than 12% followed by powdering the dried leaves to obtain powder with particles having the size below 0.42 mm;
    • b) This powder is subjected to supercritical CO2 extraction at a pressure varying between 80 Bar (80 kg/cm2) and 350 Bar (350 kg/cm2) at a temperature ranging between 31° C. to 45° C.; at a flow rate of 10 to 40 kg of CO2 per kg of raw material.
    • c) CO2 extract is separated maintaining pressure varying between 40 Bar to 65 Bar and temperature between 10° C. to 30° C. to obtain Extract A;
    • d) The remaining residual powder after separating Extract ‘A’ is subjected to further extraction using mixture of CO2 and ethyl alcohol at the pressure ranging between 80 Bar to 350 Bar and temperature ranging between 31° C. to 45° C.;
    • e) Ethyl alcohol laced with extract is collected from the separator by reducing the solvent pressure between 40 Bar and 65 Bar and temperature between 10° C. to 30° C., followed by vacuum distillation of ethanol to obtain Extract B;
    • f) Extract A and Extract B are combined, and Extract C is obtained which is called as CO2 extract of Azadirachta indica leaves.


In another aspect, the Extract A is subjected to extremely high velocity and passed through a micro-jet or nozzle to obtain fine sized nano particle extract. Nano sized delivery technologies are currently in use for sustained and enhanced delivery of active phytoconstituents. By subjecting the Extract A to high velocity micro jet, a minimum 10% of the Extract A is obtained with particle size between 10-100 nm. Hence the extract obtained by this process is considered as “Supercritical Neem Leaf Extract-Nano 10%”.


The Azadirachta indica leaves are extracted using supercritical CO2 extraction (SCO2) and under specific temperature and pressure conditions to ensure that the extract contains no harmful extraction solvents and retains the beneficiary ingredients.


Thus, supercritical CO2 extraction (SCO2) which is used in the present disclosure with or without entrained ethanol does not leave any hazardous solvent residues. The extraction temperature is maintained between 31° C. to 45° C., which ensures the retention of the temperature sensitive ingredients.


The method described herein for extraction (SCO2 extraction with or without entrained ethanol) has much lower separation temperature than the extraction temperature. The typical temperature for extraction is 31 to 45° C. while the separation temperature is around 10° C. to 30° C. in the present process. Thus, integrity of phytoconstituents is maintained along with retention of the temperature sensitive ingredients. Also, the extraction pressure is much lesser than the pressure used in the prior art (WO'199) which further ensures the retention of sensitive phytoconstituents, and thus the SCO2 extract of the present invention comprises the active components which are different from the disclosures of WO'199.


In a further embodiment, the CO2 extract thus obtained was standardized by HPLC analysis using C18 column (4.6 mm×250 mm×5 μm) and results were monitored at 215 nm using UV detection. The sample was prepared in methanol and the mobile phase was methanol and water. A gradient program sequence was used at a flow rate of 1 ml/min.


Accordingly, in another aspect, the present disclosure provides a standardized SCO2 extract which comprises nimbolide in a minimum amount of 3 mg/gm; nimbin in a minimum amount of 130 μg/gm and salinin in a minimum amount of 200 μg/gm to ensure the therapeutic efficacy of the extract when administered as the herbal formulation. The CO2 standardized extract also contain various other active phytoconstituents such as desacetylnimbin, azadiradione, azdirone, nimbolin, and nimbinene which may be contributing to this activity. However, the same are not quantified as they are in lesser quantities.


Based on the minimum concentrations of nimbolide, nimbin and nalinin in the standardized extract as mentioned above in vitro experiments were carried out using the standardized supercritical neem extract (SCNE) in human oral and colon cancer cell lines. These experiments demonstrated that the standardized CO2 extract possesses anti-proliferative activity, suppresses cancer growth and induces apoptosis through the pro-inflammatory pathway and NF-kB inhibition.


Accordingly, in another aspect, the disclosure provides herbal compositions for oral application comprising of standardized SCO2 extract of Azadirachta indica leaves in an effective amount along with one or more pharmaceutical carriers/excipients that exerts very good antitumor activity against oral and colon cancers.


Other features and advantages of the present compositions and methods are illustrated in the description below, the drawings, and the claims.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-F shows SCNE and Nimbolide inhibit oral squamous cancer cell growth. FIG. 1A shows SCNE treatment (0-400 μg/ml) for 8 h or 24 h on SCC4, HSC3, Cal27 OSCC cell lines. FIG. 1B shows SCNE treatment (0-100 μM) for 8 h or 24 h on SCC4, HSC3, Cal27 OSCC cell lines. FIG. 1C shows Nimbolide treatment (0-400 μg/ml) for 48 h on SCC4, HSC3, Cal27 OSCC cell lines. FIG. 1D shows Nimbolide treatment (0-100 μM) for 48 h on SCC4, HSC3, Cal27 OSCC cell lines. FIG. 1E shows Celecoxib treatment (0-200 μM) for 8 h or 24 h on SCC4, HSC3, Cal27 OSCC cell lines. FIG. 1F shows Celecoxib treatment (0-200 μM) for 48 h on SCC4, HSC3, Cal27 OSCC cell lines.



FIG. 2 shows SCNE and Nimbolide downregulate inflammatory mediators. SCC4, Cal27, and HSC3 cells were treated with 20 μg, 60 μg SCNE, or 10 μM, 50 μM Nimbolide for 24 h. Cytosolic protein fractions were analyzed for COX1, COX2, NFkBp65, STAT3, pSTAT3, EGFR, pEGFR, pERK1/2, AKT, and Pakt. Nuclear protein fractions were analyzed for pERK1/2, STAT3, pSTAT3, and NFkBp65. GapDH and Topo-IIα were used as loading controls.



FIGS. 3A-C shows that SCNE and Nimbolide inhibit in vitro cell migration. FIG. 3A shows using the Scratch assay that 60 μg/ml SCNE and 50 mM Nimbolide inhibits cell migration in SCC4 (120 h), Cal27 (72 h) and HSC3 (8 h). Green lines represent initial scratch front, yellow is scratch front after respective treatment time. FIG. 3B shows that the average wound width in SCC4, Cal27, and HSC3 is significantly reduced (n=6, *p<0.05, **p<0.01, ****p<0.001) with SCNE and Nimbolide treatments. FIG. 3C depicts a gelatinase zymograms showing MMP2 and MMP9 activity from SCC4, Cal27, and HSC3 treated cells.



FIG. 4A-B shows that SCNE and Nimbolide inhibit OSCC derived tumor growth in mice. FIG. 4A shows that SCC4 (30 day treatment—81.12% reduction in tumor volume), HSC3 (25 day treatment—48.81% reduction in tumor volume), and Cal27 (35 day treatment—49.00% reduction in tumor volume) cell growth was significantly inhibited in mice fed SCNE 200 mg/kg diet (*p<0.005, **p<0.001). FIG. 4B shows that Nimbolide treatment (20 mg/kg IP) significantly (*p<0.05) reduced (66%) HSC3 tumor volume in mice after 25 days.



FIGS. 5A-C shows that SCNE inhibits tumor growth in 4NQO-1 mouse model of OSCC. FIG. 6A shows that CBA mice showed no difference in weight gain on 200 mg/kg SCNE diet over the 12 week study. FIG. 5B shows that the SCNE diet reduced tongues 5 fold (**p<0.01) and tongue carcinomas compared to no SCNE diet. FIG. 5C shows that the SCNE diet reduced levels of proliferating markers PCNA, Ki-67, c-Met in mouse tongues.



FIG. 6. Shows the effects on mouse circulating cytokine levels from SCC4, Cal27, and HSC3 xenografts in mice fed SCNE.



FIG. 7 shows the effects on mouse circulating cytokine levels from a 4NQO-1 carcinogen induced model of OSCC.



FIG. 8 shows that SCNE reduces serum level inflammatory cytokines in xenografted and carcinogen induced mouse models of OSCC.



FIGS. 9A-D shows that SCNE reduced viability in CRC cells. HCT116, HT29 and IEC6 cells were treated with different concentrations of SCNE (A, B) and nimbolide (C, D) for 48 and 72 hrs and the cell viability was measured by MTT assay. Data were expressed as the mean±SD. from three independent experiments. *P<0.05 indicates significant difference versus vehicle control.



FIGS. 10A-B shows that SCNE induced apoptosis of HCT116 and HT29 cells. CRC cells were treated with SCNE (40 and 75 μg/ml) and nimbolide (5 and 10 μM) for 48 hrs. Expression of apoptosis regulatory proteins in HCT116 and HT29 cells treated with SCNE (A) and nimbolide (B) were determined by western blot analysis for Bax, Bcl-2 and Cyclin D1 with GAPDH used as a standard. Each band is representative for three experiments.



FIGS. 11A-B show that SCNE is involved in the migration of HT29 colon cancer cells. Migration scratch assays were employed to study the role of SCNE (75 μg/ml) and nimbolide (10 μM) in migration of HT29 colon cancer cells. FIG. 11A shows SCNE (75 μg/ml) and Nimbolide (10 μM) were effective in inhibiting migration of HT29 cells. FIG. 11B shows that inhibition of migration was similarly effective with the treatment of nimbolide.



FIGS. 12A-E show that inhibition of transcription factor p65 nuclear localization and STAT3 phosphorylation and pro-inflammatory markers by SCNE and nimbolide in colon cancer cells HCT116 and HT29 cells. FIG. 12A shows HCT116 and HT29 cells were treated with SCNE and nimbolide for 48 hrs and were examined by immunofluorescent staining for p65 nuclear transport. SCNE and nimbolide blocked translocation of p65 protein to the nucleus. FIG. 12B show HCT116 and HT29 cells were treated with SCNE (40 and 75 μg/ml) for 48 hrs and the expression levels of pSTAT3, p65, IKKβ and GAPDH proteins were detected by western blot analysis. FIG. 12C show HCT116 and HT29 cells were treated with nimbolide (5 and 10 μM) for 48 hrs and the expression levels of pSTAT3, p65, IKKβ and GAPDH proteins were detected by western blot analysis. The data were obtained from 3 independent experiments. FIG. 12D show HCT116 and HT29 cells were treated with SCNE (40 and 75 μg/ml) for 48 hrs and the expression levels of COX1, COX2, IL-6, and TNF-α and GAPDH proteins were detected by western blot analysis. FIG. 12E show HCT116 and HT29 cells were treated with nimbolide (5 and 10 μM) for 48 hrs and the expression levels of COX1, COX2, IL-6, TNF-α and GAPDH proteins were detected by western blot analysis. GAPDH was used as a cell internal protein marker. The data were obtained from 3 independent experiments.



FIGS. 13A-B show inhibition of invasion by SCNE and nimbolide in colon cancer cells. FIG. 13A shows HCT116 and HT29 cells were treated with SCNE (40 and 75 μg/ml) for 48 hrs and the expression levels of MMP2, MMP9 and GAPDH proteins were detected by western blot analysis. Gelatin Zymogram assay showed concentration dependent inhibition in the proteolytic activity of MMP2 on SCNE treatment. FIG. 13B show HCT116 and HT29 cells were treated with nimbolide (5 and 10 μM) for 48 hrs and the expression levels of MMP2, MMP9 and GAPDH proteins were detected by western blot analysis. Gelatin Zymogram assay showed concentration dependent inhibition in the proteolytic activity of MMP2 on nimbolide treatment.



FIG. 14 shows the dose and time dependent effect of SCNE on cell viability in normal IEC6 cells.



FIG. 15 shows the dose and time dependent effect of SCNE on cell viability in HCT 116 cells.



FIG. 16 shows the dose and time dependent effect of SCNE on cell viability in HT 29 cells



FIG. 17 shows the dose and time dependent effect of nimbolide on cell viability in HCT 116 cells



FIG. 18 shows the dose and time dependent effect of nimbolide on cell viability in HT 29 cells.



FIG. 19 shows that SCNE induces apoptosis in human colon cancer cells. HCT 116 cells were treated with varying concentration of SCNE for 72 h and DNA fragmentations were analyzed by detecting Alexa488 signal intensity.



FIG. 20 shows that SCNE increases DNA condensation in human colon cancer cells. HCT 116 cells were treated with (0-18 μg/ml) SCNE for 48 h and 72 h. Nuclei are stained blue with DAPI.



FIG. 21 shows the effect of SCNE on the cell cycle in human colon cancer cells.



FIG. 22 shows that SCNE inhibits NF-kB translocation to the nucleus in human colon cancer cells.



FIG. 23 shows that SCNE induces apoptosis in colon cancer cells. HCT116 and HT 29 cells were treated with (12-18 μg/ml) SCNE for 48 and 72 h.





DETAILED DESCRIPTION

The present disclosure can be understood more readily by reference to the following detailed description of the invention, the figures and the examples included herein.


Before the present compositions and methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, example methods and materials are now described.


Moreover, it is to be understood that unless otherwise expressly stated, it is in no way intended that any method set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not actually recite an order to be followed by its steps or it is not otherwise specifically stated in the claims or descriptions that the steps are to be limited to a specific order, it is in no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, and the number or type of aspects described in the specification.


All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided herein can be different from the actual publication dates, which can require independent confirmation.


As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.


The word “or” as used herein means any one member of a particular list and also includes any combination of members of that list.


Throughout the description and claims of this specification, the word “comprise” and variations of the word, such as “comprising” and “comprises,” means “including but not limited to,” and is not intended to exclude, for example, other additives, components, integers or steps. In particular, in methods stated as comprising one or more steps or operations it is specifically contemplated that each step comprises what is listed (unless that step includes a limiting term such as “consisting of”), meaning that each step is not intended to exclude, for example, other additives, components, integers or steps that are not listed in the step.


Ranges can be expressed herein as from “about” or “approximately” one particular value, and/or to “about” or “approximately” another particular value. When such a range is expressed, a further aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” or “approximately,” it will be understood that the particular value forms a further aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint and independently of the other endpoint. It is also understood that there are a number of values disclosed herein and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed. It is also understood that each unit between two particular units is also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.


As used herein, the terms “optional” or “optionally” mean that the subsequently described event or circumstance may or may not occur and that the description includes instances where said event or circumstance occurs and instances where it does not.


As used herein, the term “subject” refers to the target of administration, e.g., a human. Thus, the subject of the disclosed methods can be a vertebrate, such as a mammal, a fish, a bird, a reptile, or an amphibian. The term “subject” also includes domesticated animals (e.g., cats, dogs, etc.), livestock (e.g., cattle, horses, pigs, sheep, goats, etc.), and laboratory animals (e.g., mouse, rabbit, rat, guinea pig, fruit fly, etc.). In one aspect, a subject is a mammal. In another aspect, the subject is a human. The term does not denote a particular age or sex. Thus, adult, child, adolescent and newborn subjects, as well as fetuses, whether male or female, are intended to be covered.


As used herein, the term “patient” refers to a subject afflicted with a disease or disorder. The term “patient” includes human and veterinary subjects. In some aspects of the disclosed methods, the “patient” has been diagnosed with a need for treatment for cancer, such as, for example, prior to the administering step.


As used herein, the term “treating” refers to partially or completely alleviating, ameliorating, relieving, delaying onset of, inhibiting or slowing progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. Treatment can be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition. For example, the disease, disorder, and/or condition can be cancer or a hyperproliferative disorder.


As used herein, the term “inhibit” or “inhibiting” means decreasing tumor cell growth rate from the rate that would occur without treatment and/or causing tumor mass (e.g., cancer) to decrease. Inhibiting also include causing a complete regression of the tumor (e.g., cancer).


Introduction

The risk for developing oral cancers is ever increasing world-wide as the global rise in tobacco use, alcohol consumption, and HPV exposure continues (Cancer Facts and Figures 2015). Oral squamous cell carcinomas (OSCC) comprise 90% of all oral cancers and represent the 6th most common cancer in the world and the 8th most common cancer in the United States (Cancer Facts and Figures 2015). Although conventional treatments have improved the 5-year survival rates for patients with early disease, patients with late-stage disease (stage III and stage IV) have a 5-year survival rate as low as 34% (Cancer Facts and Figures 2015). Furthermore, these statistics have not changed in nearly 40 years. Hence, prevention of OSCC initiation and progression is can be important to reducing the morbidity and mortality of this devastating disease.



Azadirachta indica, or neem, belongs to a family of trees related to mahogany; meliaceae (Hao F, Kumar S, Yadav N, Chandra D. Neem components as potential agents for cancer prevention and treatment. Biochim Biophys Acta; 1846:247-57). Neem is native to India, Myanmar, Bangladesh, Sri Lanka, Malaysia, and Pakistan and grows in tropical and semi-tropical regions around the world (Hao F, Kumar S, Yadav N, Chandra D. Neem components as potential agents for cancer prevention and treatment. Biochim Biophys Acta; 1846:247-57). The neem tree is a source of highly active liminoid terpenoids, collectively known as azadiractoids which are shown to have anti-cancer activity (Manikandan P, Ramalingam S M, Vinothini G, Ramamurthi V P, Singh I P, Anandan R, et al. Investigation of the chemopreventive potential of neem leaf subfractions in the hamster buccal pouch model and phytochemical characterization. Eur J Med Chem; 56:271-81). Previous studies of neem extracts in OSCC are limited to relatively impure ethanolic organic extracts from the neem leaf evaluated in the hamster cheek pouch carcinogenesis model where some activity is shown (Subapriya R, Kumaraguruparan R, Nagini S. Expression of PCNA, cytokeratin, Bcl-2 and p53 during chemoprevention of hamster buccal pouch carcinogenesis by ethanolic neem (Azadirachta indica) leaf extract. Clin Biochem 2006; 39: 1080-7; and Dasgupta T, Banerjee S, Yadava P K, Rao A R. Chemopreventive potential of Azadirachta indica (Neem) leaf extract in murine carcinogenesis model systems. J Ethnopharmacol 2004; 92: 23-36). Murine models of stomach and skin cancers also demonstrate efficacy of the neem leaf ethanolic extract which contains at least 35 biologically active compounds (Dasgupta T, Banerjee S, Yadava P K, Rao A R. Chemopreventive potential of Azadirachta indica (Neem) leaf extract in murine carcinogenesis model systems. J Ethnopharmacol 2004; 92: 23-36).


Aerial parts and seeds of the neem tree (Azadirachta indica) have been used as medicine to treat a number of human disease conditions. Neem has a rich use in Ayurvedic traditional medicine, and its folkoric use to treat pro-inflammatory conditions has led to the hypothesis that its anti-inflammatory potential could be useful in cancer prevention and treatment. Supporting this notion is the long history of traditional use of neem to treat acute and chronic inflammatory disease in India and Africa. Neem twigs, for instance, have long been used traditionally to maintain oral health and neem has been shown to have anti-bacterial, anti-fungal, and anti-ulcer properties. These observations lend credence to the idea that neem and its constituents may modulate cancer associated inflammation.


Organic solvent extracts of neem leaf have demonstrated anti-tumor effects in models of breast, prostate, and pancreatic cancer among others. Supercritical extract technology allows for better extraction of bioactive principals from natural compounds circumventing lability of active agents to heat or to solvent degradation. In some aspects, a supercritical CO2 extract of neem leaf was used in the Examples disclosed herein and method of preparing said extract allows for better retention of innate volatiles. Neem is rich in volatile terpenoids (liminoids) which account for the bitter taste to the leaf, and as a class, are among the chief bioactive phytochemicals found in neem leaves. Few neem liminoids have been isolated in sufficient quantity to examine mechanism of action. Of the more common neem liminoids, the bulk of investigations have focused on nimbolide (5,7,4′-trihydroxy-3′,5′-diprenylflavone). Several studies have examined nimbolide for inhibition of growth from a number of cell lines, including neuroblastoma, leukemia, and melanoma. Nimbolide was also found to induce cell cycle alterations in breast and glioblastoma cell lines as well as modulate cell signaling pathways. Initial studies have reported that expression of VEGF and other metastasis enabling factors were inhibited in vitro. Supercritical leaf extracts are relatively safe to use and do not contain toxic compounds found in neem oil which is widely used as a natural insecticide. Since at least one supercritical extract of neem leaf has entered the commercial market as a constituent of many health products, its efficacy was examined in a range of colon cancer preclinical model systems, comparing its action with nimbolide.


Described herein is a supercritical CO2 extract of neem (SCNE) that is highly pure such that the bioactive component, nimbolide, has been identified and the potential solute contaminants have been removed (Rodriguez-Solana R, Salgado J M, Dominguez J M, Cortes-Dieguez S. Comparison of Soxhlet, accelerated solvent and supercritical fluid extraction techniques for volatile (GC-MS and GC/FID) and phenolic compounds (HPLC-ESI/MS/MS) from Lamiaceae species. Phytochem Anal; 26:61-71). SCNE was evaluated for anti-proliferative effects in vitro and in vivo using cell based assays, a 4NQO-1 carcinogen model of OSCC, and mouse xenograft model of human OSCC. Effects on circulating cytokines and inflammatory and apoptotic markers are also described herein in addition to disruption of EGFR signaling which drives 90% of OSCC.


Methods of Making


Azadirachta indica known as Neem is a fast-growing evergreen tree which belongs to family Meliaceae. The extracts of seeds, leaves, flowers, stem, bark and fruits of neem have consistently been used as medicine in various diseases. It is native to tropical and sub-tropical parts of India including Andhra Pradesh, Tamil Nadu and Karnataka and is also found in southeast Asia. It is also widespread in West Africa, the Caribbean and South and Central America.


The dried Azadirachta indica leaves are sourced from states of Rajastan and Madhaypradesh, India.


In this disclosure, CO2 extract derived from dried leaves of the plant is used for therapeutic application.


Accordingly, in a preferred aspect, the invention provides a process of preparing standardized CO2 extract of Azadirachta indica leaves containing beneficial phytoconstituents. The process involved following steps:

    • a) Powdering the clean and matured dried Azadirachta indica leaves having moisture to less than 12% to obtain powder with fine particles of size below 0.42 mm;
    • b) Subjecting the powder to supercritical CO2 extraction at a pressure varying between 80 Bar (80 kg/cm2) and 350 Bar (350 kg/cm2) at a temperature ranging between 31° C. to 45° C.; at a flow rate of 10 to 40 kg of CO2 per kg of raw material;
    • c) Separating CO2 extractives maintaining pressure varying between 40 Bar to 65 Bar and at a temperature lower than the extraction temperature to obtain Extract A;
    • d) Subjecting the remaining residual powder after separating Extract ‘A’ to further extraction using mixture of CO2 and ethyl alcohol at the pressure ranging between 80 Bar to 350 Bar and temperature ranging between 31° C. to 45° C.;
    • e) Collecting Ethyl alcohol laced with CO2 extract from separator by reducing the solvent pressure between 40 Bar and 65 Bar and at a temperature lower than the extraction temperature, followed by vacuum distillation of ethanol to obtain Extract B; and
    • f) Combining Extract A and Extract B to obtain Extract C which is called as CO2 standardized extract of Azadirachta indica leaves.


The matured leaves of Azadirachta indica considered for extraction are preferably of the same age.


The size of the dried powdered particles is below 0.42 mm.


The separation temperature in step c) and collection temperature in step e) is maintained between 10° C. to 30° C.


The vacuum distillation of ethanol is carried at temperature below 45° C.


The ethyl alcohol used in step d) is in an amount of 3 to 10% of the CO2.


The time required for CO2 extraction depends upon the size of the extractors and the quantity of herb loaded into the extractor at a time. The quantity of CO2 to be pumped through the herb varies between 10 kg of CO2/kg of herb to 40 kg of CO2/kg of herb depending upon the solubility of lipophilic compounds present in the herb. CO2 carries the extractives to the separator where the pressure of CO2 is reduced to a pressure varying between 40 Bar to 65 Bar and the temperature is in the range of 10° C. to 30° C. as required to separate the solute (extract) and the CO2.


This method of extraction is known as Supercritical CO2 extraction, which is the safest method of extraction for dried herbs. The extract thus obtained contains the temperature sensitive major and minor ingredients present in the herb and the other lipophilic soluble compounds. In the present invention the extract thus obtained is called as Extract A.


The remaining residual powder after isolating Extract ‘A’ is further subjected to extraction using mixture of CO2 and ethyl alcohol in proportion of 90 to 97% of Supercritical CO2 and 3 to 10% of ethyl alcohol. The extraction was carried out at the pressure ranging between 80 Bar and 350 Bar and temperature ranging between 31° C. to 45° C. The quantity of solvent pumped (CO2+Ethanol) varies between 10 kg/kg of herbs to 40 kg/kg of herbs. The solute (extract) and ethanol were separated from the CO2 on reducing the solvent pressure between 40 Bar and 65 Bar and temperature between 10° C. to 30° C. The mixture of ethyl alcohol laced with CO2 extract was collected from the separator and then subjected to vacuum distillation for separating the ethyl alcohol completely from the solute (extract). This extract is known as Extract B.


Finally, both the extracts (Extract A and Extract B) were combined to obtain Extract C. This combined extract is termed as SCO2 extract of Azadirachta indica leaves, as described in the following examples.


In another aspect, the Extract A is subjected to extremely high velocity and passed through a micro-jet or nozzle, then fine sized nano particle extract is obtained. Nano sized delivery technologies are known to potentially improve effect of the formulation. Minimum 10% of the extract is obtained with particle size between 10-100 nm. Hence the extract obtained in this aspect is considered as “Supercritical Neem Leaf Extract-Nano 10%”. Accordingly, this “Supercritical Neem Leaf Extract-Nano 10%” may be combined with extract B to obtain extract C.


The yield of the CO2 extract may be anywhere in the range of 2.5% to 5% w/w.


The supercritical CO2 extraction (SCO2) which is used in the present disclosure with or without entrained ethanol does not leave any hazardous solvent residues. The extraction temperature is maintained between 31° C. to 45° C., which ensures the retention of the temperature sensitive ingredients. Also, the extraction pressure is much lesser than the pressure of the process reported in WO'199.


The method described herein for extraction (SCO2 extraction with or without entrained ethanol) always has a much lower separation temperature than the extraction temperature. Extraction at lower temperature reduces the risk of damage to thermolabile compounds. Thus, integrity of phytoconstituents in the present disclosure is maintained apart from retaining the temperature sensitive ingredients. The typical temperature for SCF (Super Critical Fluid) extraction of the present invention is 31° C. to 45° C. while the separation temperature will be around 10° C. to 30° C.


The additional advantage of the present disclosure vis-à-vis the process reported in WO2015035199 is that the process can be achieved at much less pressure compared to the pressure of the process reported in WO'199. Moreover, the yields of the reported process in WO'199 are approximately 5%. The formulation may contain certain amount of impurities as higher extraction pressures can result in extraction of waxes and resins which remained as impurities. On contrary, the present disclosure may have lower impurities as extraction at lower pressure as in the present disclosure allows the selective extraction of active compounds.


In another embodiment, the herbal powder of Azadirachta indica leaves was subjected to water extraction to obtain the water-soluble extractives in a paste form. The extract thus obtained was dried in tray dryers/vacuum dryer or in spray drier to obtain free flowing powder extract. This extract is termed water extract (Example 5).


In another aspect, the present disclosure provides standardized SCO2 Neem extract (SCNE) which comprises nimbolide in a minimum amount of 3 mg/gm; nimbin in a minimum amount of 130 μg/gm and salinin in a minimum amount of 200 μg/gm to ensure the maximum therapeutic efficacy of the extract when administered as herbal formulation. The CO2 standardized extract also contain various other active phytoconstituents such as desacetylnimbin, azadiradione, azdirone, nimbolin, and nimbinene which may be contributing to this activity. However, the same are not quantified as they are in smaller quantities.


Accordingly, in another aspect, the disclosure provides therapeutic herbal compositions for oral application comprising of standardized SCO2 extract of Azadirachta indica leaves in an effective amount of 50 to 300 mg along with one or more pharmaceutical carriers or excipients that exerts antitumor activity against oral cancer and colon cancer.


The pharmaceutical excipients/carriers are selected from the group consisting of distilled water, saline, aqueous glucose solution, alcohol (e.g. ethanol), surfactants, propylene glycol, tween-80 and polyethylene glycol; and oily carriers such as various animal and vegetable oils, white soft paraffin, paraffin, wax, glucose, fructose, sucrose, maltose, yellow dextrin, malt dextrin, white dextrin, aerosol, aerated or fumed silica, di-calcium phosphate, microcrystalline cellulose, calcium stearate, magnesium stearate, sorbitol, stevioside, corn syrup, lactose, citric acid, tartaric acid, malic acid, succinic acid, lactic acid, L-ascorbic acid, dl-alpha-tocopherol Rosemary (Rosemarinus officinalis) CO2 extract, glycerin, propylene glycol, glycerin fatty ester, poly glycerin fatty ester, sucrose fatty ester, sorbitan fatty ester, propylene glycol fatty ester, acacia, carrageenan, casein, gelatin, pectin, agar, vitamin B group, nicotinamide, calcium pantothenate, amino acids, aerated or fumed silica, calcium salts, pigments, flavors and preservatives.


In a preferred embodiment, extract used in the formulation is made using SCO2 extraction with or without entrained solvent ethanol, using the below mentioned conditions.


Pressure of extraction: Between 72 kg/cm2 to 550 kg/cm2


Preferred Range: 80 kg/cm2 to 350 kg/cm2


Entrained ethanol: 0-10% (Preferred range 3 to 7%)


In general, the herbs are extracted using conventional methods using conventional solvents such as cold pressing method, conventional extruder press method, solvent extraction, distillation “modified atmosphere packing” (MAP).


Supercritical CO2 extraction is a process that produces an extract with a broad spectrum of the non-polar lipophilic constituent compounds present in the herb such as oils, fatty acids along with vital temperature sensitive phytonutrients. The spectrum of the extractives can be widened by using up to 10% of ethanol as an entrained solvent along with pure CO2. The major advantages of using this process of extraction are retention of the temperature sensitive ingredients present in the herb which contributes toward health benefits. Unlike other solvent extraction methods wherein solvents such as hexane, chloroform, acetone which are hazardous are used, the present extract is free of hazardous solvent residues as well as heavy metal contamination. Heavy metals are below the detectable limit in the present extract as CO2 is a highly non-polar solvent and heavy metals are highly polar, and hence they are not soluble in CO2.


When ethanol is used as entrained solvent the resulting extract is obtained by removing the ethanol from the extract to the extent under vacuum (27 to 28.5 inch of Hg) keeping temperature below 45° C., so that the residual solvent (ethanol) remains less than 1000 ppm and hence can be used safely for making the formulation.


The extract used in the present disclosure was also reduced to nanoparticles, to a size between 10 nm-100 nm, by using extremely high velocity and passing through a micro-jet or nozzle. The particles were characterized by using Dynamic Light Scattering (DLS). DLS is a light scattering technique. The basic principle of DLS is that the sample is illuminated by a laser beam and the fluctuations of the scattered light are detected at a known scattering angle by a fast photon detector. Analysis of the fluctuation of the scattered light yields information about the particles.


The SCNE thus obtained was subjected to HPLC and LC-MS to identify signature of triterpenoids with potential cancer preventive activity. Accordingly, in an additional aspect, the SCO2 Neem extract thus obtained is characterized to have minimum concentrations of nimbolide in an amount of 3 mg/gm; nimbin in an amount of 130 μg/gm and salinin in an amount of 200 μg/gm so as to ensure the efficacy of the extract prepared in the present disclosure.


In a further embodiment, the disclosure provides compositions comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE) which is characterized to comprise nimbolide, nimbin and salinin. The concentrations of any of the nimbolide, nimbin and salinin can vary in the SCNE of the present disclosure. However, in some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some other aspects, the SCNE can comprise at least 130 μg/g nimbin. In yet another aspect, the SCNE can comprise at least 200 μg/g salinin.


In some of the aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g.


In some other aspects, the composition can further comprise a pharmaceutically acceptable excipient.


Accordingly, in a further embodiment, the disclosure provides herbal pharmaceutical compositions which comprise: physiologically effective amount of the standardized (SCNE) extract with above minimum concentration of nimbolide; nimbin and salinin; or a combination thereof, in a pharmaceutical carrier/excipient to inhibit at least one of the markers of proliferation, apoptosis, and anti-cancer activity of the oral and colon cancers. The physiologically effective amount of the extract is in the range of 50 to 300 mg per day. This minimum concentration of nimbolide, nimbin and salinin or combination thereof are being achieved in the resultant SCO2 extract by using the process disclosed in this application.


The compositions can be formulated into oral solid or liquid dosage forms.


Accordingly, the therapeutically effective formulations for oral use are prepared for example, in three ways to demonstrate the present invention.


In one exemplary embodiment, the first therapeutically effective formulation for oral use contains SCO2 Azadirachta indica leaf extract: 75 mg (with minimum 0.22 mg nimbolide; 9.75 μg nimbin and salinin 15 μg); naturally occurring antioxidants such as Vitamin E (tocopherols) or Rosemary (Rosemarinus officinalis) CO2 extract containing minimum 6% carnosic acid: 10 mg and sesame oil: 415 mg; or other naturally occurring oils as a carrier. Any other suitable antioxidants can be used in place of Vitamin E or Rosemary CO2 extract. This formulation was provided as a soft gel capsule of 500 mg. The capsule can be administered to the patient 2 times a day (total 150 mg of Neem leaf extract per day as an active drug).


In another exemplary embodiment, the second therapeutically effective formulation for oral use contains SCO2 Azadirachta indica leaf extract: 50 mg (with minimum 0.15 mg nimbolide; 6.5 μg nimbin and salinin 10 μg), 582 mg dextrin/malto-dextrin or other naturally occurring carrier (e.g., di-calcium phosphate or any other suitable pharmaceutical grade carrier); and 18 mg aerated or fumed silica. A free-flowing powder was prepared and encapsulated in suitable size hard gelatin or vegetarian capsule available in the market. The same can be administered 3 times a day to get a therapeutic dose of 150 mg Neem leaf CO2 extract as described herein.


In yet another embodiment, the third therapeutically effective formulation for oral use contains SCO2Azadirachta indica leaf extract: 50 mg (with minimum 0.156 mg nimbolide; 6.5 nimbin and salinin 10 μg), 582 mg of water extract obtained from Neem leaf and 18 mg aerated or fumed silica. A free-flowing powder was prepared and encapsulated in suitable size of hard gelatin or vegetarian capsule available in the market. The same can be administered 3 times a day to get a therapeutic dose of 150-300 mg Neem leaf CO2 extract as described herein.


In another embodiment, a fine sized nano-particle CO2 extract prepared by increasing velocity and passing the particle through micro-jet or nozzle, can be administered 3 times a day to get a therapeutic dose of 100 mg Neem leaf CO2 extract nanoparticle as described herein, instead of 150 mg of ordinary CO2 extract.


In another preferred embodiment, the disclosure provides the composition in the form of a dietary supplement or an herbal medicine in conventional forms of soft gel capsules, hard gel capsules, liquid capsules, with or without carrier, dietary powders, drinks, substantially homogeneous mixture i.e., active ingredients are distributed evenly. In another preferred embodiment, the disclosure provides a pharmaceutical composition for a mouthwash which contains 4.55% of SCO2Azadirachta indica leaf extract with standard carriers and additives like sorbitol, glycerin, emulsifiers, water and suitable flavors like mint, fruits, etc. Also disclosed herein are liquid formulations. The same can be administered 3 times a day, 20 ml each time, to get a therapeutic dose of 15 mg Neem Leaf CO2 extract as described herein. The mouthwash formulation is meant for use, for example, in cases of oral cancer.


The compositions containing SCO2 Azadirachta indica leaf extract can be administered in a dosage range of 5 mg to 300 mg per dose in any of the dosage forms described herein. The recommended dose of administration is twice a day or thrice a day or two caps twice a day. In yet another embodiment, the effect of SCNE was evaluated for oral and colon cancer by measuring cell viability (by MTT assay), DNA fragmentation by TUNEL assay [Terminal deoxynucleotidyl transferase dUTP nick end labeling], DNA condensation analysis with DAPI staining, measurement of apoptotic markers (intrinsic pathway: Bax, Bcl-2) protein by Western blot analysis, Flow cytometry to study the effect of SCNE on the cell cycle were measured and the following conclusions are drawn.

    • SCNE impacts the viability of colon cancer cells in a time & dose dependent manner (FIGS. 15 to 16).
    • SCNE induces apoptosis in human colon cancer cells. HCT 116 cells were treated with varying concentration of SCNE for 72 h and DNA fragmentations were analyzed by detecting Alexa488 signal intensity (FIG. 19).
    • SCNE caused increase in DNA fragmentation. This was consistent with the profound increase in DNA condensation observed in FIG. 7.
    • Cell cycle analysis by flow cytometry confirmed a pre-apoptotic peak in SCNE treated HCT 116 cells for 48 h at IC50 (FIG. 20).
    • SCNE treatment inhibited NF-kB translocation to the nucleus (FIG. 22).
    • SCNE increased the pro-apoptotic protein Bax, and decreased the anti-apoptotic protein Bcl2 (FIG. 23).


Accordingly, in an additional embodiment, the cancer preventive effect of a super critical CO2 Neem extract of leaf (SCNE) and nimbolide alone was investigated on two human colon cancer cell lines, HCT 116 and HT 29. Normal Rat Colon cells IEC-6 cells also included in the study to verify the cytotoxicity of the SCNE extract. The effect of SCNE on cell viability was compared with nimbolide and the results are discussed herein.


The results confirm that the SCNE is non-toxic to normal Rat Colon cells IEC-6 cells even at higher concentration of 50 μg/mL after 48 hrs (FIG. 14). The SCNE treated colorectal cancer cells, viz., HCT116 and HT29 exhibits 62% (FIG. 15) and 44% cell viability (FIG. 16) respectively at a concentration of 15 μg/mL at the end of 72 hrs and exhibits zero cell viability at a concentration of 40 μg/mL (FIG. 15) and 75 μg/mL (FIG. 16) at the end of 48 and 72 hrs respectively. However, nimbolide treated colorectal cancer cells, viz., HCT116 (FIG. 17) and HT29 (FIG. 18) exhibits 80% and 75% cell viability respectively at a concentration of 15 μg/mL at the end of 48 hrs.


The experiment described herein conclusively confirmed that the super critical CO2 neem leaf extract (SCNE extract) comprising a combination of nimbolide, nimbin and salinin possess higher therapeutic efficacy than the nimbolide alone.


Compositions

Disclosed herein are compositions that can be used with any of the methods disclosed herein. The compositions described herein can be a supercritical CO2 neem extract (SCNE). Also disclosed herein are compositions comprising supercritical CO2 neem extracts. In some aspects, SCNE can comprise nimbolide, nimbin and salinin. As disclosed herein, the concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the concentrations of any of any of the components in the compositions described herein can vary.


In some aspects, the compositions described herein can further include one or more pharmaceutically acceptable excipients. Depending on the formulation, the inclusion of a pharmaceutically acceptable excipient can be optional. Examples of pharmaceutically acceptable excipients that can be used include but are not limited to dextrin/malto-dextrin or di-calcium phosphate, distilled water, saline, aqueous glucose solution, alcohol (e.g. ethanol), surfactants, propylene glycol, tween-80 and polyethylene glycol; and oily carriers such as various animal and vegetable oils, white soft paraffin, paraffin, wax, glucose, fructose, sucrose, maltose, yellow dextrin, malt dextrin, white dextrin, aerosol, microcrystalline cellulose, calcium stearate, magnesium stearate, sorbitol, stevioside, corn syrup, lactose, citric acid, tartaric acid, malic acid, succinic acid, lactic acid, L-ascorbic acid, dl-alpha-tocopherol, glycerin, propylene glycol, glycerin fatty ester, poly glycerin fatty ester, sucrose fatty ester, sorbitan fatty ester, propylene glycol fatty ester, acacia, carrageenan, casein, gelatin, pectin, agar, vitamin B group, nicotinamide, calcium pantothenate, amino acids, aerated or fumed silica, calcium salts, pigments, flavors and preservatives.


In some aspects, the SCNE can comprise one or more liminoids. In some aspects, the composition further comprises one or more tocopherols; and sesame oil. Examples of tocopherols include but are not limited to alpha-tocopherol, gamma-tocopherol, vitamin E (tocopherols) or Rosemary (Rosemarinus officinalis), CO2 extract or any o anti-oxidant which is pharmaceutically accepted. In some aspects, the composition further comprises one or more tocopherols; sesame oil; and aerated or fumed silica.


In some aspects, the composition can comprise SNCO2 extract: 75 mg; antioxidants such as, for example, Vitamin E or Rosemary (Rosemarinus officinalis) CO2 extract: 10 mg and sesame oil: 415 mg.


In some aspects, the composition can comprise SNCO2 extract: 50 mg; 582 mg dextrin/malto-dextrin or other carrier (e.g., di-calcium phosphate or any other pharmaceutical grade carrier); and 18 mg aerated or fumed silica.


In some aspects, the composition can comprise water extract obtained from Azadirachta indica leaf and used as a carrier for making a free-flowing powder of 50 mg Neem leaf extract obtained with supercritical CO2 extraction as described herein.


In some aspects, the composition can comprise SNCO2 extract: 2.28 g, peppermint (Mentha piperita) oil: 13.81 g, spearmint (Mentha spicata) oil: 9.26 g, clove bud (Syzigium aromaticum) CO2 oil: 3.98 g, tween 80: 20.68 g. 1.25 g of the said blend was diluted in 98.75 g of Base. The base comprises: water: 73.5 g, Aloe Vera water (200×): 10 g, sorbitol: 10 g, glycerin: 5.9 g, ascorbic acid: 0.5 g, potassium sorbate: 0.1 g.


Methods of Treatment

Disclosed herein, are methods of treating cancer in a subject, the method comprising: (a) identifying a subject in need of treatment; and (b) administering to the subject a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In an aspect, the SCNE can comprise nimbolide, nimbin and salinin. Also, disclosed herein, are methods of treating cancer in a subject, the method comprising: (a) identifying a subject in need of treatment; and (b) administering to the subject a therapeutically effective amount of a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In some aspects, SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the concentrations of any of any of the components in the compositions described herein can vary. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human.


Disclosed herein, are methods of reducing at least one inflammatory cytokine in serum of a subject in need thereof. In some aspects, the method can comprise administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In an aspect, the SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human. In some aspects, the at least one inflammatory cytokine can be IFN-γ, TNF-α, IL-6 or IL-1. In some aspects, the subject can have, be suspected of having or be diagnosed with oral cancer. In some aspects, the administration to the subject of a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract, wherein SCNE comprises nimbolide, nimbin and salinin can reduce at least one inflammatory cytokine in serum of the subject, wherein the subject has, is suspected of having or has been diagnosed with oral cancer. In some aspects, the at least one inflammatory cytokine can be IL-6 or TNF-α. In some aspects, IFN-γ, TNF-α, IL-6 or IL-1 can be reduced after the administration of a therapeutically effective amount of a supercritical CO2 neem extract described herein in a subject that has, is suspected of having, or has been diagnosed with oral cancer, In some aspects, the subject's serum can have increased levels of at least one inflammatory cytokine when compared to a reference sample before the administration of the composition comprising therapeutically effective amount of a supercritical CO2 neem extract. In some aspects, the methods can further comprise determining the level of at least one inflammatory cytokine in one or more cells of a subject before the administration of the composition comprising a therapeutically effective amount of a supercritical CO2 neem extract. In some aspects, the level of at least one inflammatory cytokine can be higher when compared to a reference sample.


Disclosed herein, are methods of reducing inflammation in a subject in need thereof. The method can comprise administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In some aspects, the SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human. In some aspects, the subject has been diagnosed with oral cancer or colon cancer prior to the administering step. In some aspects, the inflammation can be reduced by decreasing the expression of one or more of IFN-γ, IFN-β, TNF-α, IL-6, IL-1, NF-KB, STAT3, COX1 or COX2.


Disclosed herein, are methods of treating a hyperproliferative disorder in a subject in need thereof. The method can comprise administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In an aspect, the SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human. In some aspects, the subject has been diagnosed with a need for treatment of the hyperproliferative disorder prior to the administering step. In an aspect, the hyperproliferative disorder can be cancer. In some aspects, the hyperproliferative disorder can be oral cancer or colon cancer.


Disclosed herein, are methods of suppressing expression of NFkB and cycloxygenase in a subject in need thereof. The method can comprise administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In some aspects, the SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human. In some aspects, the subject has been diagnosed with a need for suppressing the expression of NFkB and cyclooxygenase prior to the administering step. In some aspects, the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step. In some aspects, the methods can further comprise the step of identifying a subject in need of treatment of a disorder of uncontrolled cellular proliferation. In some aspects, the disorder of uncontrolled cellular proliferation can be a cancer. In some aspects, the cancer can be an oral cancer.


Disclosed herein, are methods of suppressing expression of NFkB and cyclooxygenase in at least one cell. In some aspects, the method can comprise the step of contacting at least one cell with an effective amount of a supercritical CO2 neem extract (SCNE). In some aspects, SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In some aspects, the at least one cell can be a human cell. In some aspects, the contacting step can be via administration to a subject. In some aspects, the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step. In some aspects, the subject has, is suspected of having or has been diagnosed with oral cancer. In some aspects, the uncontrolled cellular proliferation can be oral cancer.


Disclosed herein, are methods of modifying epidermal growth factor receptor signaling (EGFR) activity in a subject. The method can comprise administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In some aspects, the SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human. In some aspects, the subject has been diagnosed with a need for modifying EGFR signaling activity. In some aspects, the modifying can be inhibiting. In some aspects, SCNE can inhibit EGFR signaling activity. In some aspects, the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step. In some aspects, the methods further comprise the step of identifying a subject in need of treatment of a disorder of uncontrolled cellular proliferation. In some aspects, the disorder of uncontrolled cellular proliferation can be oral cancer.


Disclosed herein, are methods of inducing apoptosis of a cell in a subject in need thereof. The method can comprise administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE). In some aspects, the SCNE can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In some aspects, the composition can further comprise a pharmaceutically acceptable excipient. In an aspect, the subject can be a human. In some aspects, subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step. In some aspects, the methods can further comprise the step of identifying a subject in need of treatment of a disorder of uncontrolled cellular proliferation. In some aspects, the disorder of uncontrolled cellular proliferation can be a cancer. In some aspects, the cancer can be colon cancer.


The compositions described herein can be formulated to include a therapeutically effective amount of a supercritical CO2 neem extract described herein. In an aspect, the supercritical CO2 neem extract can comprise nimbolide, nimbin and salinin. The concentrations of any of nimbolide, nimbin and salinin can vary. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. Therapeutic administration encompasses prophylactic applications. Based on genetic testing and other prognostic methods, a physician in consultation with their patient can choose a prophylactic administration where the patient has a clinically determined predisposition or increased susceptibility (in some cases, a greatly increased susceptibility) to a type of cancer.


The concentration or amounts of each of nimbolide, nimbin and salinin can vary in a single composition. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. The concentration or amount of each component can vary according to many factors, for example, the particular the type and severity of the cancer as well as the type of formulation.


In some aspects, the SCNE can comprise one or more liminoids. In some aspects, the SCNE further comprises azadirachtin A, azadirachtin B, and deacetylsalinin. In some aspects, the SCNE further comprises small amounts of one or more of azadirachtin A, azadirachtin B, and deacetylsalinin. In an aspect, “small amounts” can refer to amounts that can be at the limit of detection.


In some aspects, the compositions described herein can further comprise one or more tocopherols; and sesame oil. Examples of tocopherols include but are not limited to alpha-tocopherol, gamma-tocopherol, vitamin E (tocopherols) or Rosemary (Rosemarinus officinalis), CO2 extract or any other naturally occurring anti-oxidant which is pharmaceutically accepted. In some aspects, the composition further comprises one or more tocopherols; sesame oil; and aerated or fumed silica.


The compositions described herein can be administered to the subject (e.g., a human patient) in an amount sufficient to delay, reduce, or preferably prevent the onset of clinical disease. Accordingly, in some aspects, the patient can be a human patient. In therapeutic applications, compositions are administered to a subject (e.g., a human patient) already with or diagnosed with cancer in an amount sufficient to at least partially improve a sign or symptom or to inhibit the progression of (and preferably arrest) the symptoms of the condition, its complications, and consequences. An amount adequate to accomplish this is defined as a “therapeutically effective amount.” A therapeutically effective amount of a composition (e.g., a pharmaceutical composition) can be an amount that achieves a cure, but that outcome is only one among several that can be achieved. As noted, a therapeutically effective amount includes amounts that provide a treatment in which the onset or progression of the cancer is delayed, hindered, or prevented, or the cancer or a symptom of the cancer is ameliorated. One or more of the symptoms can be less severe. Recovery can be accelerated in an individual who has been treated.


Disclosed herein, are methods of treating a patient with cancer. The cancer can be any cancer. In some aspects, the cancer can be oral cancer or colon cancer. In some aspects, the cancer can be a primary or secondary tumor. In an aspect, the subject has been diagnosed with cancer prior to the administering step.


The compositions described herein can be formulated to include a therapeutically effective amount of supercritical CO2 neem extract as described herein alone or in combination with one or more therapeutic agents or therapies or treatment regimens. In an aspect, the one or more therapeutic agents or therapies or treatment regimens can be chemotherapy or radiotherapy. In an aspect, SCNE can be contained within a pharmaceutical formulation. In an aspect, the pharmaceutical formulation can be a unit dosage formulation. The compositions described herein can be formulated in a variety of combinations. The particular combination of SCNE with one or more chemotherapeutic agent or radiotherapy can vary according to many factors, for example, the particular the type and severity of the cancer.


The therapeutically effective amount or dosage of the supercritical CO2 neem extract as described herein used in the methods as disclosed herein applied to mammals (e.g., humans) can be determined by one of ordinary skill in the art with consideration of individual differences in age, weight, sex, other drugs administered and the judgment of the attending clinician. Variations in the needed dosage may be expected. Variations in dosage levels can be adjusted using standard empirical routes for optimization. The particular dosage of a pharmaceutical composition to be administered to the patient will depend on a variety of considerations (e.g., the severity of the cancer symptoms), the age and physical characteristics of the subject and other considerations known to those of ordinary skill in the art. Dosages can be established using clinical approaches known to one of ordinary skill in the art.


The duration of treatment with any composition provided herein can be any length of time from as short as one day to as long as the life span of the host (e.g., many years). For example, the compositions can be administered once a day; once a week (for, for example, 4 weeks to many months or years); once a month (for, for example, three to twelve months or for many years); or once a year for a period of 5 years, ten years, or longer. It is also noted that the frequency of treatment can be variable. For example, the present compositions can be administered once (or twice, three times, etc.) daily, weekly, monthly, or yearly. In an aspect, the compositions described herein can be administered twice to three times a day. In some aspects, the compositions described herein can be administered twice to three times a day for two to three to four weeks (or longer). In some aspects, the compositions described herein can be administered two to three times a day. In some aspects, the compositions described herein can be administered two to three times a day for two, three or four weeks.


Dosages of SCNE can be in the range of 50 mg to 1000 mg/day. In an aspect, SCNE can be administered in a dosage ranging from about 50 mg to 1000 mg/day. In an aspect, the dosage of SCNE can be 25, 50, 75, 100, 125, or 150 mg/day or any amount in between. In some aspects, the dosage of SCNE can be more than 150 mg/day. In some aspects, the dosage of SCNE can be 160, 170, 180, 190, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950 or 1000 mg/day or any amount in between. In an aspect, the compositions described herein can be in the form of a capsule. In some aspects, the capsule can be administered orally one, two or three times a day. In some aspects, the compositions can be administered orally one, two or three times a day for 21 or 28 days. In some aspects, the compositions can be administered orally two or three times a day for 21 or 28 days. In some aspects, the compositions can be administered for 1 month, 2 months, 3 months, 4 months, 5 months or 6 months or longer. In some aspects, the can be administered orally two or three times a day for 6 months.


In some aspects, the total effective amount of the compositions as disclosed herein can be administered to a subject as a single dose, either as a bolus over a relatively short period of time, or can be administered using a fractionated treatment protocol in which multiple doses are administered over a more prolonged period of time.


In some aspects, the compositions described herein can be administered in conjunction with other therapeutic modalities to a subject in need of therapy. The present compounds can be given to prior to, simultaneously with or after treatment with other agents or regimes. For example, supercritical CO2 neem extract as described herein can be administered in conjunction with standard therapies used to treat cancer. In an aspect, any of the compositions described herein can be administered or used together with chemotherapy or radiotherapy.


Pharmaceutical Compositions

As disclosed herein, are pharmaceutical compositions, comprising supercritical CO2 neem extract as described herein and a pharmaceutical acceptable carrier described herein. In some aspects, SCNE can be formulated for oral administration. The compositions can be formulated for administration by any of a variety of routes of administration, and can include one or more physiologically acceptable excipients, which can vary depending on the route of administration. As used herein, the term “excipient” means any compound or substance, including those that can also be referred to as “carriers” or “diluents.” Preparing pharmaceutical and physiologically acceptable compositions is considered routine in the art, and thus, one of ordinary skill in the art can consult numerous authorities for guidance if needed.


In some aspects, the compositions disclosed herein can be administered directly to a subject. Generally, the compositions can be suspended in a pharmaceutically acceptable carrier (e.g., physiological saline or a buffered saline solution) to facilitate their delivery. Encapsulation of the compositions in a suitable delivery vehicle (e.g., polymeric microparticles or implantable devices) may increase the efficiency of delivery.


In some aspects, the compositions can be formulated in various ways for parenteral or nonparenteral administration. Where suitable, oral formulations can take the form of tablets, pills, capsules, or powders, which may be enterically coated or otherwise protected. Sustained release formulations, suspensions, elixirs, aerosols, and the like can also be used. In an aspect, the composition can be in a form comprising a capsule.


Pharmaceutically acceptable carriers and excipients can be incorporated (e.g., water, saline, aqueous dextrose, and glycols, oils (including those of petroleum, animal, vegetable or synthetic origin), starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monosterate, sodium chloride, dried skim milk, glycerol, propylene glycol, ethanol, and the like). In some aspects, the pharmaceutically acceptable excipient can be dextrin/malto-dextrin or di-calcium phosphate. In some aspects, the excipient can vary depending on the formulation. In some aspects, the excipient can be optional. In some aspects, the pharmaceutically acceptable excipient and carrier can be selected from the group consisting of distilled water, saline, aqueous glucose solution, alcohol (e.g. ethanol), surfactants, propylene glycol, tween-80 and polyethylene glycol; and oily carriers such as various animal and vegetable oils, white soft paraffin, paraffin, wax, glucose, fructose, sucrose, maltose, yellow dextrin, malt dextrin, white dextrin, aerosol, microcrystalline cellulose, calcium stearate, magnesium stearate, sorbitol, stevioside, corn syrup, lactose, citric acid, tartaric acid, malic acid, succinic acid, lactic acid, L-ascorbic acid, dl-alpha-tocopherol, glycerin, propylene glycol, glycerin fatty ester, poly glycerin fatty ester, sucrose fatty ester, sorbitan fatty ester, propylene glycol fatty ester, acacia, carrageenan, casein, gelatin, pectin, agar, vitamin B group, nicotinamide, calcium pantothenate, amino acids, aerated or fumed silica, calcium salts, pigments, flavors and preservatives. The compositions may be subjected to conventional pharmaceutical expedients such as sterilization and may contain conventional pharmaceutical additives such as preservatives, stabilizing agents, wetting or emulsifying agents, salts for adjusting osmotic pressure, buffers, and the like. Suitable pharmaceutical carriers and their formulations are described in “Remington's Pharmaceutical Sciences” by E. W. Martin, which is herein incorporated by reference. Such compositions will, in any event, contain an effective amount of the compositions together with a suitable amount of carrier so as to prepare the proper dosage form for proper administration to the patient.


The pharmaceutical compositions as disclosed herein can be prepared for oral administration. In an aspect, the composition can be administered orally. Pharmaceutical compositions prepared for parenteral administration include those prepared for intravenous (or intra-arterial), intramuscular, subcutaneous, intraperitoneal, transmucosal (e.g., intranasal, intravaginal, or rectal), or transdermal (e.g., topical) administration. Aerosol inhalation can also be used. Thus, compositions can be prepared for parenteral administration that includes SCNE dissolved or suspended in an acceptable carrier, including but not limited to an aqueous carrier, such as water, buffered water, saline, buffered saline (e.g., PBS), and the like. One or more of the excipients included can help approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents, and the like. Where the compositions include a solid component (as they may for oral administration), one or more of the excipients can act as a binder or filler (e.g., for the formulation of a tablet, a capsule, and the like). In an aspect, the compositions can be for the formulation of a liquid (e.g., a mouthwash). The pharmaceutical compositions can be sterile and sterilized by conventional sterilization techniques or sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation, which is encompassed by the present disclosure, can be combined with a sterile aqueous carrier prior to administration. The pH of the pharmaceutical compositions typically will be between 3 and 11 (e.g., between about 5 and 9) or between 6 and 8 (e.g., between about 7 and 8). The resulting compositions in solid form can be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules. In an aspect, the compositions can be packaged in a container comprising multiple doses in a liquid form.


In an aspect, a pharmaceutical composition comprises SCNE; and optionally, a pharmaceutical acceptable carrier. The SCNE can comprise nimbolide, nimbin and salinin. In some aspects, the SCNE can comprise at least 3 mg/g nimbolide. In some aspects, the SCNE can comprise at least 130 μg/g nimbin. In some aspects, the SCNE can comprise at least 200 μg/g salinin. In some aspects, the amount of the nimbolide present in the composition can be at least 3 mg/g; the amount of the nimbin present in the composition can be at least 130 μg/g nimbin; and the salinin present in the composition can be at least 200 μg/g. In an aspect, the pharmaceutical composition can be formulated for oral administration. In an aspect, the composition can be formulated as a capsule or a liquid.


In some aspects, the compositions described herein can be provided in a therapeutically effective formulation for oral administration comprising: SNCO2 extract: 75 mg; antioxidants such as, for example, Vitamin E (tocopherols) or Rosemary (Rosemarinus officinalis) CO2 extract: 10 mg and sesame oil: 415 mg. Said composition can be put into a soft gel capsule of 500 mg. In some aspects, the capsules can be administered to a patient two times a day for a total of 50-1000 mg to four times a day for a total of 1000 mg of Neem leaf extract per day as an active drug.


In some aspects, the compositions described herein can be provided in a therapeutically effective formulation for oral administration comprising: SNCO2 extract: 50 mg; 582 mg dextrin/malto-dextrin or other carrier (e.g., di-calcium phosphate or any other pharmaceutical grade carrier); and 18 mg aerated or fumed silica. A free-flowing powder can be prepared and encapsulate in “00” size hard gelatin or vegetarian capsule. In an aspect, the treatment regimen can be 1 capsule twice a day to 3 capsules thrice a day in 21 or 28-days cycle for 6 cycles. In some aspects, the compositions described herein can be provided in a therapeutically effective formulation for oral administration comprising: water extract obtained from Azadirachta indica leaf and used as a carrier for making a free-flowing powder of 50 mg Neem leaf extract obtained with supercritical CO2 extraction as described herein. Said composition can be administered two times a day to achieve a therapeutic dose of 150 mg to four times a day for a total of 1000 mg Neem leaf CO2 extract as described herein.


In some aspects, the compositions described herein can be provided in a therapeutically effective formulation as a mouthwash comprising: SNCO2 extract: 2.28 g, peppermint (Mentha piperita) oil: 13.81 g, spearmint (Mentha spicata) oil: 9.26 g, clove bud (Syzigium aromaticum) CO2 oil: 3.98 g, tween 80: 20.68 g. 1.25 g of the said blend was diluted in 98.75 g of Base. The base comprises: water: 73.5 g, Aloe Vera water (200×): 10 g, sorbitol: 10 g, glycerin: 5.9 g, ascorbic acid: 0.5 g, potassium sorbate: 0.1 g. The formulation described herein is a liquid formulation that can be administered 1-3 times a day, 20 ml each time, to achieve a therapeutic dose of 150-1000 mg Neem Leaf CO2 extract as described above. The mouthwash formulation can be used to treat or prevent oral cancer.


In some aspects, a fine-sized nano-particle CO2, in which a minimum of 10% of the extract are nanoparticles, can be prepared by increasing the velocity and passing the particle through a micro-jet or nozzle, can be administered orally 2 or 3 times a day to achieve therapeutically effective dose of 100-500 mg Neem leaf CO2 extract nanoparticle as described above, instead of 1000 mg of ordinary CO2 extract.


The formulations disclosed herein can be prepared using the nano-particle extracts described herein.


Articles of Manufacture

The composition described herein can be packaged in a suitable container labeled, for example, for use as a therapy to treat cancer or any of the methods disclosed herein. Accordingly, packaged products (e.g., sterile containers containing the composition described herein and packaged for storage, shipment, or sale at concentrated or ready-to-use concentrations) and kits, including at least SCNE as described herein and instructions for use, are also within the scope of the disclosure. A product can include a container (e.g., a vial, jar, bottle, bag, or the like) containing the composition described herein. In addition, an article of manufacture further may include, for example, packaging materials, instructions for use, syringes, buffers or other control reagents for treating or monitoring the condition for which prophylaxis or treatment is required. The product may also include a legend (e.g., a printed label or insert or other medium describing the product's use (e.g., an audio- or videotape)). The legend can be associated with the container (e.g., affixed to the container) and can describe the manner in which the compound therein should be administered (e.g., the frequency and route of administration), indications therefor, and other uses. The compounds can be ready for administration (e.g., present in dose-appropriate units), and may include a pharmaceutically acceptable adjuvant, carrier or other diluent. Alternatively, the compounds can be provided in a concentrated form with a diluent and instructions for dilution.


EXAMPLES
Example 1: Oral Squamous Cell Carcinoma Growth Inhibition by Neem (Azadirachta indica) Extract In Vivo: Disruption of the Inflammation Cascade, Reduction in Tumor Occurrence and Volume in Oral Squamous Cell Carcinoma by Treating with a Natural Neem Leaf Extract

The leaves and bark of the Neem tree (Azadirachta indica) have been used in traditional Ayurvedic medicine for centuries to treat oral maladies. The experiments described herein tested the hypothesis that use of this Neem leaf extract can prevent initiation and/or progression of OSCC. The anti-cancer potential of the Neem leaf extract was tested in in vitro and in vivo platforms. OSCC cell lines (SCC4, Cal27, HSC3) were treated with the leaf extract at various time points while markers of inflammation, invasion, and proliferation were analyzed. The preventive effects of SCNE were also assessed in ectopic xenograft mouse models and a carcinogen induced mouse model of oral cancer. Treatment with the Neem leaf extract inhibited OSCC cell proliferation, reduced levels in markers of inflammation in OSCC cells. The Neem leaf extract reduced wound closure, showing inhibition of metastasis. Xenografted nude mice showed significant reduction of OSCC tumor occurrence and reduced tumor growth. The Neem leaf extract also significantly reduced tumors and tongue dysplasias in a 4NQO-1 mouse oral carcinogen model. In both cancer animal models, the Neem Leaf extract depressed circulating inflammatory cytokines. Chemopreventive effects of SCNE were also examined on the inhibition and prevention of OSCC both in vitro and in vivo. The results show a marked decrease in tumor proliferation, reduction of inflammatory markers and circulating cytokines, which strongly support the potential of SCNE as prevention agent in a standalone regime or in combination with standard frontline therapies to improve patient outcomes.


Materials and Methods

Reagents.


Supercritical CO2 neem extract was provided by Nisarga Ltd, Sartara, India. Leaves from organically grown neem trees are processed with supercritical CO2. Supercritical extracts have the advantage of replacing organic solvents with excellent solvency and no organic residues remain resulting in a highly pure neem extract (SCNE) (Lindskog M A, Nelander H, Jonson A C, Halvarsson T. Delivering the promise of SFC: a case study. Drug Discov Today; 19: 1607-12). Stock solutions of 50 mg/ml in 100% DMSO were used in vitro. Nimbolide was purchased from Biovision (#2356) and dissolved in 100% DMSO to a stock of 1 mg/ml. SCNE diets were manufactured by Harlan Teklad to deliver 200 mg/kg SCNE—SNCE was dissolved in corn oil and homogenously mixed with the remaining diet ingredients and formed into pellets. Celecoxib (PZ0008, Sigma, USA) was dissolved in 100% DMSO to a stock of 1 mg/ml.


Human OSCC Cell Lines and MTT Assay.


SCC4 and Cal27 oral cancer cells are obtained from ATCC and maintained in DMEM supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin at 37° C. with 5% CO2. For the SCC4 cells, hydrocortisone was provided at 400 ng/ml in the completed media. For the cell treatments, SCNE (Nisarga Ltd.) was used and applied at different concentrations (1-400 μg/m) for 8 h, 24 h, or 48 h to 75% confluent cells. Doses to be tested will bracket the IC50s for each cell line. Control cells will receive DMSO. Nimbolide was applied at different concentrations (1-100 μM) for 8 h, 24 h, or 48 h to 50% confluent cells. For the celecoxib treatments cell were treated at different concentrations (1-200 ug/ml) for 8 h, 24 h, or 48 h. Cells were cultured overnight in complete media, serum-starved for 24 hr, and treated vehicle, SCNE, nimbolide, or Celecoxib as described herein. Subsequently, 10 μl of 12 mM MTT (Life Technologies; Carlsbad, Calif.) solution was added to each well, incubated for 4 hr at 37° C., and neutralized with DMSO. Absorbance was measured at 540 nm and percent viability was calculated.


Gelatinase Zymogram.


Gelatinase zymography was performed in 10% SDS polyacrylamide gel in the presence of 0.1% gelatin under non-reducing conditions. Colon cancer cells were grown in 96 well plates. Culture media 200 μl was collected from each well (pool of 3×) and concentrated to final volume 20 Culture media (20 μl) were mixed with sample buffer and loaded for SDS-PAGE without boiling. Following electrophoresis the gels were washed twice in 1× Zymogram renaturing Buffer containing Triton X-100 (Thermo Scientific, MA) for 1 hr at Room Temperature to remove SDS. The gels were then incubated in 1× Zymogram Developing Buffer containing the substrate (Thermo Scientific, MA) for 48 hrs at 37° C. and stained with 0.5% Coomassie Blue R250 in 50% methanol and 10% glacial acetic acid for 60 min and destained. Upon renaturation of the enzyme, gelatinases digest gelatin in the gel and give clear bands against an intensely stained background. Protein standards and 2% fetal bovine serum (positive control) were run concurrently and appropriate molecular weights were determined by plotting the relative mobilities of known proteins (PMID 28440509).


Cell Migration Assay.


SCC4, Cal27, and HSC3 cells were cultured in 96-well plates in complete growth medium. A scratch was performed using a WoundMaker and visualized using the IncuCyte ZOOM real time imaging system (Essen BioScience, MI, USA). Cells were treated with 20 or 60 μg/ml SCNE or 10 or 50 μM Nimbolide and imaged at 3 hr intervals for 72-120 hr to monitor cell migration and wound healing.


Protein Expression.


Cellular protein extracts will be prepared and proteins quantified as described previously. Briefly, cells will be washed twice with 1×PBS, collected by scraping and centrifuged at 4° C. at 300 g for 6 min. The pellet will be resuspended in 250 μl of Buffer A (10 mM Tris-HCl pH 7.8, 10 mM KCl, 1.5 mM MgCl2, 1 tab protease inhibitor and water) and incubated on ice for 10 min. The samples will then be homogenized at 15,000 rpm for 45 s on ice and then centrifuged at 4,600 g for 5 min at 4° C. The supernatent will be removed and stored in −80° C. as the cytosolic protein fraction. The collected pellet will be resuspended in 100 μl of Buffer B (210 mM Tris-HCl pH 7.8, 420 mM KCl, 1.5 mM MgCl2, 20% glycerol, 1 tab of protease inhibitor and water) followed by gentle agitation at 4° C. for 30 min and centrifugation at 10,000 g for 10 min at 4° C. The supernatant will be collected and stored at −80° C. as the nuclear protein fraction. Fifty micrograms of either cytosolic or nuclear protein fractions will be separated by SDS-PAGE (12% gels) and transferred to PVDF membranes (Bio-Rad, USA). The membranes will be probed with the primary antibodies (Cell Signalling, USA) NFkB p65 (8242S), STAT3 (8768S) pSTAT3 (9131S), COX1 (9896S), COX2 (12282S), EGFR (4267S), pEGFR (4404S), ERK1/2 (T202/Y204—9101S), AKT (9272S), pAKT (9271S) followed by horseradish peroxidase-conjugated anti-rabbit (7074S). GAPDH (2118S) or Topo IIα (12286S) will be used to ensure equal protein loading. The immunoreactive bands will be visualized on ChemiDoc Touch (Bio-Rad, USA) using chemiluminescent substrate (Clarity ECL, Bio-Rad). Bands will be quantitated with ChemiDoc software (Bio-Rad).


Animals.


Six week-old female athymic nude mice (Harlan, Indianapolis, Ind.) were used in a laminar air-flow cabinet under pathogen-free conditions. They were provided with a 12 hr light/dark schedule at controlled temperature and humidity with food and water ad libitum. Mice were acclimated for one week prior to study initiation.


OSCC Mouse Xenograft Models.


Mice were injected subcutaneously in the right flank with 3×106 HSC3 or 10×106 SCC4 or 6×106 Cal27 cells in 0.2 ml of sterile PBS as previously described. Mice were placed on AIN76A synthetic diet for 24 hours. Then SCNE diet (200 mg/kg) was placed into the Neem treatment group, then control group remained on the AIN76A. For the HSC3 animal groups, nimbolide was injection by intraparentaerally for 5 consecutive days, starting at day 10 post tumor injection, at 5 mg or 20 mg Nimbolide/kg mouse. Tumor volumes were calculated by the elliptical formula: ½(Length×Width2) (Jensen M M, Jorgensen J T, Binderup T, Kjaer A. Tumor volume in subcutaneous mouse xenografts measured by microCT is more accurate and reproducible than determined by 18F-FDG-microPET or external caliper. BMC Med Imaging 2008; 8:16). Blood was drawn at termination and serum isolated for cytokine analysis.


CBA Carcinogen Induced Oral Cancer Model.


Twenty CBA mice were placed on AIN76A or 200 mg/kg SCNE diet and given 4-NQO-1 (Sigma) at 100 μg/ml in their drinking water. The mice were kept on the 4NQO-1 water for 8 weeks, followed by 4 weeks of regular water. At 12 weeks, blood was drawn at terminations for serum cytokine analysis and tongues excised and fixed in formalin.


Immunohistochemistry.


The formalin fixed tongue were paraffin embedded and sliced at 1 microns. Immunostaining was done following method previously published (PMID 27167203) using the following antibodies (Abcam: PCNA ab18197; Ki-67 ab16667; c-Met ab51067).


Cytokine and Chemokine Assay.


Serum cytokine/chemokine profiles were taken at termination and stored at −80° C. until analysis using the Bio-Plex Pro group 1 mouse cytokine 23-plex assay kit and analyzed with the Bio-Plex 200 Luminex-based multiplex analysis system (Bio-Rad, Hercules, Calif.).


Statistical Analysis.


Statistical analysis was performed using GraphPad Prism4 (San Diego, Calif.). Cell viability and migration assays were analyzed by one-way ANOVA and Bonferroni's post-hoc test. Statistical analyses of tumor growth were made using analysis of variance with repeated measures with Bonferroni's post-hoc test. A p value less than 0.05 was considered statistically significant.


Results

SCNE and Nimbolide Inhibit Oral Squamous Cancer Cell Growth.


There are previous reports of some anti-cancer of Neem extracts, mainly alcohol derived, as well as, effects with Nimbolide, a single compound from in Neem leaf extracts. However, to date there are no reports investigating the anti-cancer potential of a solvent free, hydrophobic and hydrophilic constituent containing, supercritical CO2 extract of Neem leaves. Utilizing SCNE and Nimbolide, three different OSCC human cell lines were treated at various doses and three time points (8 h, 24 h, 48 h) to determine cytotoxic concentrations and IC50 (FIG. 1A-D). The SCNE reduced cell growth in a dose and time dependent manner with an IC50 of 50 μg/ml of SNCE at the time points tested (FIG. 1A, 1C) and 15 μM Nimbolide for three time points (FIG. 1B, 1D). Next, the SCNE and Nimbolide cytotoxic effects were compared to a standard non-steroidal anti-inflammatory, Celecoxib (FIG. 1E-F). At 8 h and 24 h treatments the IC50 for Celecoxib was 75 μM as well as a similar IC50 at 48 h was markedly less than Nimbolide. These results show that SCNE and Nimbolide have cytotoxic effects on OSCC cell lines similar to or slightly higher than standard NSAIDs. From this data, 20 and 60 μg/ml SCNE and 10 and 50 μM were chosen to further understand the mechanisms of action.


SCNE and Nimbolide Down Regulate Inflammatory Mediators.


To elucidate the mechanism(s) of action of SCNE and Nimbolide, three OSCC cell lines were treated with 20 μg, 60 μg/ml SCNE and 10 mM, 50 mM Nimbolide and analyzed for cytosolic and nuclear protein fractions (PMID 27167203). Reports have shown that inflammatory markers, such as NFkB, cyclooxygenases, as well as cellular proliferators STAT3, AKT, and ERK1/2 are elevated in OSCC. Treatment with SCNE or Nimbolide moderately decreases COX2 levels with minimal effect on COX1 (FIG. 2) but a modest effect on NFkBp65 at higher doses of SCNE and Nimbolide was observed. Both SNCE and Nimbolide showed drastic down regulation of pSTAT3, pAKT and pERK1/2. However, little change in EGFR and pEGFR was observed in response to treatments. In the nuclease, SCNE and Nimbolide showed stronger reduction on NFkBp65 and pERK1/2. A similar trend in STAT3 and pSTAT3 reduction was also observed in response to SCNE and Nimbolide. These results confirm the anti-inflammatory and anti-proliferative potency of SCNE and Nimbolide in OSCC.


SCNE and Nimbolide Inhibit In Vitro Cell Migration.


The in vitro results suggests a strong cytotoxic effect on OSCC through down-regulation of inflammatory mediators and cellular proliferation markers. To better understand the anticancer potential of SCNE and Nimbolide, their anti-metastatic effects were assessed. Utilizing a wound healing assay, it was observed that both SCNE and Nimbolide significantly reduce cell migration (FIG. 3A-B). The highly mobile cell line HSC3 reduced the wound (90%) in 8 h, however, SCNE and Nimbolide inhibited this closure, with less than 10% closure. In the less mobile line, SCC4, SCNE and Nimbolide halted cell migration across the wound in a similar fashion, although this occurred after 120 h (FIGS. 3A-B). Cal27 cells are not very mobile, however, SNCE and Nimbolide did inhibit the modest cell migration compared to the untreated group. Given these strong results in perturbing cellular migration, the effects of SCNE and Nimbolide were assessed on two metalloprotease proteins MMP2 and MMP9. In the highly mobile HSC3 OSCC cell line, SNCE and Nimbolide reduced MMP2 activity with slight reduction in MMP9. In the SCC4 cell line, MMP9 was drastically reduced SCNE and Nimbolide, with little change in MMP2. The non-mobile Cal27 cell line had modest reduction in MMP2 activity with SCNE treatment and MMP9 treatment with Nimbolide. Taken together, the in vitro results suggest a strong anti-tumor effect of SCNE and Nimbolide through down regulation of proliferative markers, reduction in inflammatory markers, and reduced cellular migration.


SNCE and Nimbolide Inhibits OSCC Derived Tumor Growth in Mice.


To corroborate the results from the OSCC experiments, the same three cell lines in xenografted mouse models (FIG. 4) were used. 200 mg/kg SCNE was incorporated into the diet to deliver the therapeutic neem extract and a synthetic AIN73A diet as control. In one cell line, HSC3, 5 mg or 20 mg Nimbolide was injected by IP for 5 consecutive days. At termination the SCNE diet significantly reduced SCC4 tumor volume (81%) and Cal27 (49%) volume compared to the untreated controls (FIG. 4A). SCNE did reduce HSC3 tumor volume (49%), however, high variance did not produce a significant result. The 20 mg/kg Nimbolide treatment did significantly reduce tumor volume (69%) and the 5 mg/kg showed a moderate reduction (40%) in volume (FIG. 4B). Body weight data were comparable between experimental arms (data not shown). These data affirm both SCNE and nimbolide have profound anti-tumor activity in vivo.


SCNE and Nimbolide Reduce Serum Level Inflammatory Cytokines in Xenografted Mice.


The serum from the above mice used to investigate the effects of SCNE on the circulating inflammatory cytokine population (FIG. 8) was analyzed. The extract strongly reduced IL-1b, TNFα, IFNγ, and IL-6 serum levels. For IL-1α, there was a moderate reduction in the level of IL-1α identified in the xenografted mice. A similar pattern was observed for IL-10 but the stronger reduction occurred in the HSC3 tumor bearing mice. Many other cytokines from these animals were examined (FIG. 6) and overall SCNE treatment produced a different profile compared to control. Combined with tumor volume reduction, the data suggests that SCNE can reduce tumor burden and deleterious inflammatory cytokines.


SCNE Inhibits Tumor Growth in 4NQO-1 Mouse Model of OSCC.


To further validate the in vivo anti-cancer potential of SCNE, a 4-NQO-1 induced tongue OSCC model in the CBA strain was established. The 4NQO-1 (50 μg/ml) was administered in the drinking water for 8 weeks and then regular water replaced it for another 4 weeks. The control mice were fed ad libitum AIN73A diet and the treatment group the same 200 mg/kg SCNE diet for the entire 12 week study. To assess the palatability of the diet, mice weight was measured every 2 weeks and the mice showed no difference in weight gain (FIG. 5A). At termination, the tongues pathobiology was examined for any dysplasias and/or tumors. The SCNE significantly reduced early stage dysplasia compared to controls and 66% reduction in OSCC tumors (FIG. 5B). The expression levels of proliferative markers in the tongues was also characterized by immunohistochemistry and it was found that SCNE reduced PCNA, Ki-67, and c-Met protein levels (FIG. 5C).


SCNE Reduces Serum Inflammatory Cytokines in a Carcinogen Induced Mouse Model of OSCC.


In addition to the pathobiology of these mice, the serum circulating cytokine inflammatory population was examined. The SCNE significantly reduced IFNγ, IL-1β and TNFα levels in these animals (FIG. 8). Two other cytokines IL-6 (30%) and IL-1α (25%) levels were reduced after 12 weeks of SCNE diet consumption. Many other cytokines from these animals were examined (FIG. 7) and overall SCNE and Nimbolide treatment produced a different profile compared to control. This pattern follows similar reductions in the xenograft animal studies further suggesting the anti-cancer and anti-inflammatory effects of SCNE.


Discussion

The anti-cancer effects of SCNE in OSCC was shown to occur through downregulation of key tumor proliferating markers and reduction in inflammatory modulators. The Neem leaf extract reduced wound closure, showing inhibition of metastasis. Xenografted nude mice showed significant reduction of OSCC tumor occurrence and reduced tumor growth. The Neem leaf extract also significantly reduced tumors and tongue dysplasia in an 4NQO-1 mouse oral carcinogen model. In both cancer animal models, the Neem Leaf extract depressed circulating inflammatory cytokines.


Currently, OSCC cancers are on the rise and in the clinic most chemotherapies are of the standard variety with very little second line options. To improve this scenario, new avenues to treatment with novel agents and combination therapies could overcome this problem. For instance, COX2 expression has been shown to be elevated in OSCC cases and contribute to radioresistance.


The chemo preventive effects of SCNE, known for its biomedicinal properties, were examined on the inhibition and prevention of OSCC, both in vitro and in vivo. The data described herein show a marked decrease in tumor proliferation, reduction of inflammatory markers, and circulating cytokines. With the dearth of clinical treatment options for OSCC frontline and second line therapies, this extract may be used as a prevention agent in a standalone regime or in combination with standard frontline therapies to improve patient outcomes and/or resistant recurrent tumors in relapsed patients.


Example 2: A Supercritical CO2 Extract of Neem Leaf (A. indica) and its Bioactive Liminoid, Nimbolide, Suppress Colon Cancer in Preclinical Models by Modulating Pro-Inflammatory Pathways

To explore a role of neem in CRC, human colon cancer cell lines HCT116 and HT29 cells were treated with purified Super Critical Neem Extract (SCNE) or nimbolide. SCNE treatment showed a dose dependent inhibition of CRC cell proliferation and an increase in apoptosis. It was found that treatment of both SCNE and nimbolide showed anti-inflammatory effect by poor nuclear localization of p65, decreased protein expression of transcriptional factor phosphorylated STAT3 and pro inflammatory cytokines COX1, COX2, IL-6 and TNF-α in CRC cells. Western blots and Zymogram results showed anti-invasive effect by decreased expression of MMP2 and MMP9 proteins in CRC cells on treatment with SCNE. Overall, these data confirms a potential anti-cancer effect of SCNE, reducing cell proliferation, inflammation, migration, and invasion and inducing apoptosis in human Colon cancer cells.


Materials and Methods

Cell lines and Cell Culture.


Human colon cancer cell lines HCT116 and HT29 were obtained from American Type Culture Collection (ATCC). Both of these cell lines were cultured in McCoy's 5A medium supplemented with pyruvate, vitamins, amino acids, antibiotics and 10% fetal bovine serum. Rat colon normal epithelial cell line IEC6 was obtained from American Type Culture Collection [IEC6] (ATCC® CRL-1592™). IEC6 Cells were cultured in Dulbecco's modified Eagle's medium (DMEM) with 4 mM L-glutamine, 1.5 g/L sodium bicarbonate, 4.5 g/L glucose, 0.1 Unit/ml bovine insulin and 10% fetal bovine serum. The cultures were maintained at 37° C. in a humidifier incubator with 5% CO2. To determine dose-dependent changes in protein and gene expression, cells were treated with different concentrations of Supercritical Extract Neem Extract, SCNE (Nisarga, India) and nimbolide (Biovision, USA) or an equal volume of Dimethyl Sulfoxide (DMSO) as a vehicle for different time period as needed.


Cell Viability Assay.


Colorectal cancer cells, HCT116 and HT29 as well as normal Rat Colon cells IEC-6 cells, were plated in 96-well plates, next day the cells were serum starved for 24 hrs and treated with SCNE (0-150 μg/mL) and nimbolide (1-100 μM) for 48 h and 72 h. After treatment, cell viability was measured by MTT [3-(4, 5-Dimethylthiazol-2-yl)-2, 5-Diphenyltetrazolium Bromide] assay (Sigma Aldrich, MO) according to the manufacturer's instructions. Briefly, MTT (5 mg/mL) was added and plates were incubated at 37° C. for 4 h before dimethyl sulfoxide was added to each well. Finally, the absorbance of each well was read at a wavelength of 540 nm using a plate reader (Molecular Devices, Sunnyvale, Calif., USA). The results were expressed as a percentage of surviving cells over non treated cells.


Migration Scratch Assay.


Migration assays were performed using the IncuCyte ZOOM system (Essen BioScience, Inc., MI) to measure migration of the colon cancer cells without and with SCNE and nimbolide treatment. A wound was created in confluent cells in each well (10 replicates) and the ingrowth of the cells were calculated over a definite time period by masking wound boundaries at 0 h against untreated cells, to measure wound closure.


Western Blot Analysis.


HCT-116 and HT-29 cells were grown to confluent in 100 mm culture dishes. Cells were serum starved for 24 hrs. Next day, the cells were treated with different doses of SCNE and nimbolide against vehicle (DMSO) for 48 hrs at 37° C. Whole cell lysates were prepared with RIPA lysis buffer. Nuclear Protein extraction: Non-treated and treated HCT-116 and HT-29 cells were kept on ice for 10 min with low salt Lysis buffer (10 mM HEPES, 10 mM KCl, 1 mM EDTA), then scrapped and spun down. Pellet was collected and 50-100 μl of high-salt lysis buffer was added and incubated on ice for 30 min with intermediate vortex. The tubes were spun down and nuclear protein was collected from the supernatant. Protein concentration was determined using Pierce BCA Protein Assay Kit (Thermo Scientific, MA).


Equal amounts of protein were separated on 7.5%, 10% and 12% SDS PAGE. Then, proteins were transferred to Immun-Blot PVDF membranes for protein Blotting (Bio Rad, CA) and blocked in 5% non-fat milk in Tris-buffered saline with 0.1% Tween-20 (TBST) for 1 h at room temperature. Antibodies against COX1 (Cell Signaling Technology, MA), COX2 (Cell Signaling Technology, MA), Bcl-2 (Abcam, MA), Bax1 (Abcam, MA), TNF-α COX2 (1:500; Cell Signaling Technology, MA), IL-6 (Cell Signaling Technology, MA), Cyclin D1 (Cell Signaling Technology, MA), p65 (Abcam, MA), IKKβ (Abcam, MA), MMP2 (Abcam, MA), MMP9 (Abcam, MA), pSTAT3 (Y705) (Cell Signaling Technology, MA), Topoisomerase, (Abcam, MA), and GAPDH (Sigma-Aldrich, MO) were diluted in 5% skim milk. Horseradish peroxidase-conjugated goat anti-rabbit (Abcam, MA) antibody was used as a secondary antibody.


Gelatinase Zymogram:


Gelatinase zymography was performed in 10% SDS polyacrylamide gel in the presence of 0.1% gelatin under non-reducing conditions. Colon cancer cells were grown in 96 well plates. Culture media 200 μl was collected from each well (pool of 3×) and concentrated to final volume 20 μl. Culture media (20 μl) were mixed with sample buffer and loaded for SDS-PAGE without boiling. Following electrophoresis the gels were washed twice in 1× Zymogram renaturing Buffer containing Triton X-100 (Thermo Scientific, MA) for 1 hr at Room Temperature to remove SDS. The gels were then incubated in 1× Zymogram Developing Buffer containing the substrate (Thermo Scientific, MA) for 48 hrs at 37° C. and stained with 0.5% Coomassie Blue R250 in 50% methanol and 10% glacial acetic acid for 60 min and destained. Upon renaturation of the enzyme, the gelatinases digest the gelatin in the gel and give clear bands against an intensely stained background. Protein standards and 2% fetal bovine serum (positive control) were run concurrently and appropriate molecular weights were determined by plotting the relative mobilities of known proteins (25997494).


Immunofluorescence Microscopy:


Quiescent human CRC cells were grown in multiwell plastic chamber slides and treated with SCNE or nimbolide for 48 hrs. At the termination of the study time, cells were washed twice with ice-cold PBS and fixed in methanol at −20° C. for 5 min. After a brief rinse, cells were blocked with 0.1% BSA in PBS and then stained with p65 using indirect immunofluorescence. Alexa Fluor 594 donkey anti-rabbit antibody served as secondary antibody (Thermo Fisher Scientific, MA). Stained cells were washed with PBS, mounted with prolong Gold antifade reagent with DAPI (Thermo Fisher Scientific, MA) mounted with coverslips, viewed and photographed using Zeiss LSM710 Confocal microscope (Carl Zeiss Microscopy, LLC, NY).


Results

SCNE and Nimbolide Inhibited Proliferation in Human CRC Cells.


To evaluate the effect of SCNE on human CRC cells, cell viability was analyzed using MTT assay. To investigate whether SCNE and Nimbolide have direct effects on CRC cells, the proliferation inhibition caused by SCNE and Nimbolide was tested in HCT-116 and HT-29 human CRC cell lines as well as normal rodent colon cell line IEC-6 by the MTT assay. Treatment of HCT-116 and HT29 with different concentrations of SCNE and Nimbolide for 48 and 72 h, resulted in a decrease in the cell viability (FIG. 9). Normal rodent colon cell line growth (IEC6) was not affected by SCNE as well as nimbolide. These results show that SCNE could inhibit CRC cell viability in a concentration and time dependent manner. The IC50 for the SCNE and Nimbolide were determined to be <75 μg/ml and <10 μM, respectively. In further experiments, CRC cells were treated with SCNE dose of 40 μg/ml and 75 μg/ml and nimbolide dose of 5 μM and 10 μM for 48 hrs.


SCNE Induced Apoptosis in CRC Cells.


Suppression of apoptosis during carcinogenesis is thought to play a central role in the development and progression of some cancers. Tumor cells can acquire resistance to apoptosis by the expression of either anti-apoptotic proteins such as Bcl-2 or by down-regulation of pro-apoptotic proteins such as Bax. To figure out the relationship between induction of apoptosis and the expression of their regulatory proteins by SCNE treatment, the expression of apoptosis regulatory proteins was investigated. SCNE resulted in a decreased expression of anti-apoptotic marker Bcl-2 protein and upregulation of pro-apoptotic marker Bax protein in both HCT116 and HT29 cells (FIG. 10A) which was similar to nimbolide treatment in HCT116 and HT29 cells (FIG. 10B).


Cyclin D1 is a protein required for progression through the G1 phase of the cell cycle. Overexpression of Cyclin D1 has been shown to correlate with early cancer onset and tumor progression. CRC cell lines showed higher expression of Cyclin D1 protein which was significantly reduced upon treatment with SCNE and Nimbolide for 48 hrs (FIG. 10).


SCNE Inhibited Migration of Human CRC Cells.


It was then determined whether the cell anti-proliferative and apoptotic activity of the SCNE and Nimbolide might translate into possible inhibition of cell migration, forecasting a potential inhibition of invasion. To test this, migration assays were conducted using the IncuCyte ZOOM system to measure migration of CRC cells without and with SCNE and Nimbolide treatment. It was found that SCNE and Nimbolide, both inhibited wound closure after 72 h treatment in a dose-dependent manner in HT-29 human colon cancer cells (FIG. 11).


SCNE have Anti-Inflammatory Activity—NF-κB/IL-6/STAT3 Expression in CRC Cells.


NF-κB and STAT3 regulate the expression of a large number of genes involved in inflammation. To determine whether SCNE and Nimbolide treatment on CRC cell lines HCT-116 and HT-29 exhibited an anti-inflammatory effect, the impact of SCNE and Nimbolide treatment on CRC cell lines was assessed. Here, it was found that treatment of HCT-116 and HT-29 cells for 48 h with the IC50 of SCNE and Nimbolide resulted in reduced translocation of p65 from cytoplasm to the nucleus (FIG. 12A, 12B, 12C) and decreased expression of pSTAT3 protein expression, showing a loss of available NF-κB and STAT3 transcriptional factors to the nucleus.


IL-6 and TNF-α are pro-inflammatory cytokines and are highly expressed during cancer. The results from the experiments described herein show that treatment of CRC cell lines with SCNE and Nimbolide remarkably decreased the expression of IL-6 and TNF-α protein (FIG. 12D, 12E).


COX1 is constitutively expressed in human colon tissues whereas tumorigenic factor such as COX2 has been involved in colon tumorigenesis. The results showed that treatment of SCNE and Nimbolide decreased the protein expression of both COX1 and COX2 in CRC cell lines (FIG. 12D, 12E). Taken together, these data showed that SCNE and Nimbolide have an anti-inflammatory effect on CRC cell lines.


SCNE Inhibited Invasion in Human CRC Cells.


MMPs are involved in invasion, migration, metastasis and tumorigenesis. Among the many MMPs that have been identified, gelatinases, especially MMP-2 (gelatinase A) and MMP-9 (gelatinase B), are thought to a play a key role in degradation of type IV collagen and gelatin, the two main components of ECM. To examine that metalloproteinase was responsible, gelatinase Zymography of HCT116 and HT29 cells treated with SCNE and Nimbolide against vehicle was performed. HCT116 and HT29 cells demonstrated strong secretion of MMP2 in serum free media which was inhibited by SCNE and Nimbolide after 48 h of treatment (FIG. 13A, 13B). More MMP2 expression in the medium directly correlated to more digestion of gelatin in the gel resulting in a clear band in untreated cells. Western blot analysis of human CRC cell lines showed higher expression of MMP2 and MMP9 in untreated cells. Treatment of colorectal cancer cells with SCNE and Nimbolide significantly decreased the expression of MMPs.


Discussion

Given the high mortality rate as a result of colon cancer and the significant morbidity, apparent toxicity and poor response rates of current chemotherapeutic regimens, there has been a big push to identify novel therapeutic modalities with fewer toxicity profiles. Targeted therapies against VEGF (bevacizumab) or against EGFR (cetuximab) are now commonly used as treatments for CRC. However, patients develop resistance to such treatments; thus, new strategies are required to replace or complement current therapies. Dietary alterations can lead to widespread differences in the risks and incidences of several types of cancers. Additionally, the long term consumption of natural products present in fruits and spices, with proven safety, favoring their use in cancer chemoprevention. The approach of tumor prevention using safe and nontoxic novel plant derived agents has been fortified by many scientists. Plentiful natural products have been investigated for their potential use as anticancer agents. Neem is one such natural herb with demonstrable anti-cancer properties and is a source of several limonoids, which are a class of oxygenated triterpenes called tetranorterpenoids. These limonoids are responsible for the anti-tumor effects of neem leaf extract (NLE). However, the underlying mechanism of its inhibition of colorectal cancer cell proliferation and metastasis remains to be elucidated.


As described herein, it was investigated whether SCNE can exert anticancer activity against CRC via modulation of proinflammatory pathways in CRC cells and in animal model. Cell viability was assessed using an MTT assay in the absence or presence of various concentrations of SCNE. It was found that SCNE inhibited proliferation, migration and induced apoptosis in CRC cells. Therefore, the anti-proliferative and anti-migratory effects of SCNE observed in the present study were dependent on its cancer-preventive effects. Nimbolide caused cell cycle arrest at G1/S phase. Evidently, nimbolide was found to reduce cyclin A level, which is required for colon cancer cells to proceed through S phase, hence inducing cell cycle arrest and resulting in inhibition of cell growth. Antiapoptotic proteins and proapoptotic proteins regulate the level of activation of caspase3. Nimbolide treatment decreased the expression of antiapoptotic proteins (Bcl-xL, Bcl-2, survivin, caspase inhibitor molecules) and increased the expression of proapoptotic proteins (cytochrome c, Bax, Bad, Bid, cleaved caspases) in prostate cancer cells, which is similar to the results disclosed herein with SCNE treatment in CRC cells. The overexpression of proteins associated with cell survival and cell proliferation has been shown to contribute to tumor development. Down-regulation of the expression of proteins involved with cell survival and proliferation may contribute to the decreased growth of colon cancer cells. The observed antiproliferative and apoptosis inducing properties of SCNE are in agreement with those observed by others in leukemia and in colon cancer.


In the present study, the results described herein show that SCNE can inhibit the expression of proteins involved in tumor invasion, metastasis, and angiogenesis (MMP-9, MMP-2) which further supports the role of SCNE against CRC. Fucoidan inhibited cell growth, migration and sphere formation by suppressing the PI3K/Akt/mTOR pathway and reducing the expression of MMP-2 in human HT-29 colon cancer cells. Magnolol significantly downregulated matrixmetalloproteinase-9 (MMP9) expression, an enzyme critical to tumor invasion and also inhibited nuclear factor-kB (NF-kB) transcriptional activity showing that its role in suppresses tumor invasion by inhibiting MMP-9 through the NF-kB pathway in human breast cancer. (24226295). Nimbolide inhibited proliferation, induced apoptosis, and suppressed NF-κB activation and NF-κB-regulated tumorigenic proteins in CRC cells. Nimbolide injected intraperitoneally after tumor inoculation, significantly decreased the volume of CRC xenografts. The limonoid-treated xenografts exhibited significant down-regulation in the expression of proteins involved in tumor cell survival (Bcl-2, Bcl-xL, c-IAP-1, survivin, Mcl-1), proliferation (cMyc, cyclin D1), invasion (MMP-9, ICAM-1), metastasis (CXCR4), and angiogenesis (VEGF).


It was found that HCT-116 and HT-29 colon cancer cells exhibited constitutive NF-κB and that SCNE suppressed this activation. It was shown that nimbolide inhibited inducible and constitutive NF-κB activation in leukemia and multiple myeloma cells. Constitutive NF-κB has been found to be important for the survival and proliferation of various tumor cell types by regulating the expression of proteins involved in tumor development. Therefore, it is likely that nimbolide exerts its inhibitory effects on tumor survival and growth by inactivating NF-κB. One of the possible mechanisms for the constitutive activation of NF-κB in tumor cells is through IKK activation. Avicins were found to be potent inhibitors of TNF-α induced NF-κB and to slow the accumulation of the p65 subunit of NF-κB in the nucleus. Avicin G treatment decreased the expression of NF-κB regulated proteins such as iNOS and COX-2. Other studies showed that pretreating cells with triterpenoids for 24 hours significantly reduced the induction of NF-κB mediated through TNF-α. Cycloartane triterpenoids from Cimicifuga dahurica suppressed the expression of cdc2 and COX-2 protein. These results imply that triterpenoids possess potential antitumor activities and exert their cytotoxicity through apoptosis and G2/M cell cycle arrest. Nimbolide was found to inhibit IκB degradation and prevent nuclear translocation of NF-κB. This subsequently caused cell cycle arrest by downregulating numerous genes involved in cellular proliferation. Nimbolide can induce apoptosis through inactivation of NF-κB. This led to significant suppression of Bcl-2 with concomitant increase in the expression of Bax, cytochrome C, and Smac/DIABLO (Kavitha 2012).


The results described herein, however, are the first to demonstrate the potential of the SCNE in inhibiting the growth of CRC cells and in a xenograft nude mouse model. It was found that SCNE can mediate antitumor activity in vivo by modulating the expression of numerous tumorigenesis-related proteins. First, SCNE down-regulated the expression of Bcl-2 which is known to promote tumor survival. Second, SCNE down-regulated the expression of cyclin D1 which is known to be overexpressed in CRC and to promote tumor growth. Third, SCNE down-regulated the expression of proteins involved in tumor invasion, metastasis, and angiogenesis such as MMP-9 and MMP2. Fourth, constitutively active NF-κB and STAT3 known to regulate the expression of all of these proteins, was also inhibited by the SCNE treatment.


REFERENCES



  • Arsene D, Galais M P, Bouhier-Leporrier K, Reimund J M. Recent developments in colorectal cancer treatment by monoclonal antibodies. Expert Opin Biol Ther. 2006; 6:1175-92.

  • Babykutty S, Priya P S, Nandini R J, Kumar M A S, Nair M S, Srinivas P and Gopala S. Nimbolide retards tumor cell migration, invasion, and angiogenesis by downregulating MMP-2/9 expression via inhibiting ERK1/2 and reducing DNA-binding activity of NF-kappa B in colon cancer cells. Molecular Carcinogenesis. 2012; 51(6):475-490.

  • Deeb, D.; Gao, X.; Jiang, H.; Dulchaysky, S. A.; Gautam, S. C. Oleanane triterpenoid CDDO-Me inhibits growth and induces apoptosis in prostate cancer cells by independently targeting pro-survival Akt and mTOR. Prostate 2009, 69, 851-860.

  • Fakih M. Anti-EGFR monoclonal antibodies in metastatic colorectal cancer: time for an

  • Greten F R, Eckmann L, Greten T F, Park J M, Li Z W, Egan L J, et al. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004; 118:285-96.

  • Gupta S C, Prasad S, Reuter S, Kannappan R, Yadav V R, Ravindran J, et al. Modification of cysteine 179 of IkappaBalpha kinase by nimbolide leads to down-regulation of NF-kappaB-regulated cell survival and proliferative proteins and sensitization of tumor cells to chemotherapeutic agents. J Biol Chem. 2010; 285:35406-17.

  • Gupta S C, Reuter S, Phromnoi K, Park B, Hema P S, Nair M, et al. Nimbolide sensitizes human colon cancer cells to TRAIL through reactive oxygen species- and ERK-dependent up-regulation of death receptors, p53, and Bax. J Biol Chem. 2011; 286:1134-46.

  • Gupta Subhash C, Sandeo Prasad, Dhanya R. Sethumadhavan, Mangalam S. Nair, Yin-Yuan Mo and Bharat B. Aggarwal. Nimbolide, a Limonoid Triterpene, Inhibits Growth of Human Colorectal Cancer Xenografts by Suppressing the Proinflammatory Microenvironment. Clin Cancer Res. 2013; 19(16): 4465-4476. doi:10.1158/1078-0432.CCR-13-0080.

  • Jackson-Bernitsas D G, Ichikawa H, Takada Y, Myers J N, Lin X L, Darnay B G, et al. Evidence that TNF-TNFR1-TRADD-TRAF2-RIP-TAK1-IKK pathway mediates constitutive NF-kappaB activation and proliferation in human head and neck squamous cell carcinoma. Oncogene. 2007; 26:1385-97.

  • Han Yong-Seok, Lee Jun Hee, Lee Sang Hun. Fucoidan inhibits the migration and proliferation of HT-29 human colon cancer cells via the phosphoinositide-3 kinase/Akt/mechanistic target of rapamycin pathway. Molecular medicine reports, 12: 3446-3452, 2015. DOI: 10.3892/mmr.2015.3804.

  • Haridas, V.; Arntzen, C. J.; Gutterman, J. U. Avicins, a family of triterpenoid saponins from Acacia victoriae (Bentham), inhibit activation of nuclear factor-kappaB by inhibiting both its nuclear localization and ability to bind DNA. Proc. Natl. Acad. Sci. USA 2001; 98 (20):11557-62.

  • Fakih M. Anti-EGFR monoclonal antibodies in metastatic colorectal cancer: time for an individualized approach? Expert Rev Anticancer Ther. 2008; 8:1471-80. [PubMed: 18759698]

  • Karin M, Cao Y, Greten F R, Li Z W. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002; 2:301-10.

  • Kavitha K, Priyadarsini R V, Anitha P, Ramalingam K, Sakthivel R, Purushothaman G, Singh A K, Karunagaran D and Nagini S. Nimbolide, a neem limonoid abrogates canonical NF-kappa B and Wnt signaling to induce caspasedependent apoptosis in human hepatocarcinoma (HepG2) cells. European Journal of Pharmacology. 2012; 681(1-3):614.

  • Kumar HarishG, Vidya Priyadarsini R, Vinothini G, Vidjaya Letchoumy P, Nagini S. The neem limonoids azadirachtin and nimbolide inhibit cell proliferation and induce apoptosis in an animal model of oral oncogenesis. Invest New Drugs. 2010; 28:392-401.

  • Mermelshtein A, Gerson A, Walfisch S, Delgado B, Shechter-Maor G, Delgado J, et al. Expression of D-type cyclins in colon cancer and in cell lines from colon carcinomas. Br J Cancer. 2005; 93:338-45.

  • Park W, Amin A R, Chen Z G, Shin D M (2013) New perspectives of curcumin in cancer prevention. Cancer Prev Res (Phila) 6: 387-400.

  • Singh P R, Arunkumar R, Sivakamasundari V, Sharmila G, Elumalai P, Suganthapriya E, Mercy A B, Senthilkumar K and Arunakaran J. Anti-proliferative and apoptosis inducing effect of nimbolide by altering molecules involved in apoptosis and IGF signalling via PI3K/Akt in prostate cancer (PC-3) cell line. Cell Biochemistry and Function. 2014; 32(3):217-228.

  • Tian, Z.; Yang, M.; Huang, F.; Li, K.; Si, J.; Shi, L.; Chen, S.; Xiao, P. Cytotoxicity of three cycloartane triterpenoids from Cimicifuga dahurica. Cancer Lett. 2005, 226 (1): 65-75.

  • Ying Liu, Wei CaoBo Zhang, Yong-qiang Liu, Zhong-yuan Wang, Yan-ping Wu, Xian-jun Yu, Xu-dong Zhang, Ping-hong Ming, Guang-biao Zhou, Laiqiang Huang. The natural compound magnolol inhibits invasion and exhibits potential in human breast cancer therapy. Scientific Reports 2013; 3:3098|DOI: 10.1038/srep03098.

  • Youns M, Efferth T, Hoheisel J D (2009b) Microarray analysis of gene expression in medicinal plant research. Drug Discov Ther 3: 200-207.

  • Youns M, Efferth T, Reichling J, Fellenberg K, Bauer A, Hoheisel J D (2009a) Gene expression profiling identifies novel key players involved in the cytotoxic effect of Artesunate on pancreatic cancer cells. Biochem Pharmacol 78: 273-283. doi: 10.1016/j.bcp.2009.04.014

  • Youns M, Hoheisel J D, Efferth T (2010) Toxicogenomics for the prediction of toxicity related to herbs from traditional Chinese medicine. Planta Med 76: 2019-2025. doi: 10.1055/s-0030-1250432

  • Paul R, Prasad M, Sah N K. Anticancer biology of Azadirachta indica L (neem): a mini review. Cancer Biol Ther 2011; 12: 467-476.



Example 3: CO2 Leaf Extract of Azadirachta indica

One KG of cleaned and matured Azadirachta indica leaves were taken and dried under the shade to reduce moisture content to less than 12%. It was ensured that the powder contains moisture less than 12%. Such dried herb was powdered in particle size below 0.42 mm and then subjected to SCO2 extraction at a pressure varying between 80 Bar (80 kg/cm2) and 350 Bar (350 kg/cm2) at a temperature ranging between 31° C. and 45° C. The CO2 was passed through the herb for a period of 2-3 hours depending upon the size of the extractors and the quantity of herb loaded into the extractor at a time. The quantity of CO2 to be pumped through the herb varies between 10 kg of CO2/kg of herb to 40 kg of CO2/kg of herb depending upon the solubility of lipophilic compounds present in the herb. The CO2 carried extractives were collected from the separator where the pressure of CO2 was reduced to a pressure varying between 40 Bar to 65 Bar and temperature between 10° C. to 30° C. to separate the solute (extract) and the CO2. The extract thus obtained contains the temperature sensitive ingredients present in the herb and the other lipophilic soluble compounds. The extract thus obtained is Extract A.


Residual powder after isolating Extract ‘A’ was subjected to extraction using mixture of CO2 and ethyl alcohol in proportion of 90 to 97% of Supercritical CO2 and 3 to 10% of ethyl alcohol. The extraction was carried out at the pressure ranging between 80 Bar and 300 Bar and temperature ranging between 31° C. to 45° C. The quantity of solvent pumped (CO2+Ethanol) varies between 10 kg/kg of herbs to 40 kg/kg of herbs. The solute (extract) and ethanol were separated from the CO2 on reducing the solvent pressure between 40 Bar and 65 Bar and temperature between 10° C. to 30° C. The ethyl alcohol laced with extract was collected from the separator. The mixture was then subjected to vacuum distillation (27 to 28.5 inch of Hg) keeping temperature below 45° C., for separating the ethyl alcohol completely from the solute (extract), which was named as Extract B.


Residual solvent (ethanol): less than 1000 ppm


Both the extracts extract ‘A’ and extract ‘B’ were combined to obtain Extract C.


Yield: 2.5-5%


Example 4: Water Extract of Azadirachta indica

The herb powder of Azadirachta indica leaves (1 KG) was subjected to water extraction to obtain the water-soluble extractives in a paste form. The extract thus obtained was dried in tray dryers/vacuum dryer or in spray dryer to obtain free flowing powder extract. This extract is termed as water extract.


Yield: 5:1%.


Example 5: Standardization of the CO2 Extract

The standardization of the CO2 extract was carried out using HPLC. C18 column (4 mm×250 mm×5 μm) was used. The sample was prepared in methanol and the mobile phase was methanol and water. A gradient program sequence was used where the run time was 60 minutes and the flowrate were 1 ml/min. The extract obtained is having a minimum of nimbolide in an amount of 3 mg/gm, nimbin in an amount of 130 μg/gm and salinin in an amount of 200 μg/gm.


Example 6: Therapeutically Effective Formulations for Oral Use

Formulation 1.


SCO2 Azadirachta indica leaf extract: 75 mg (with minimum 0.22 mg nimbolide; 9.75 μg nimbin and salinin 15 μg); antioxidants such as Vitamin E (tocopherols) or Rosemary (Rosemarinus officinalis) CO2 extract containing minimum 6% carnosic acid: 10 mg and sesame oil: 415 mg; this formulation was filled in a soft gel capsule of 500 mg.


This capsule can be administered to the patient 2-4 times a day (total 150-300 mg of Neem leaf extract per day as an active drug).


Formulation 2.


SCO2 Azadirachta indica leaf extract: 50 mg (with minimum 0.15 mg nimbolide; 6.5 μg nimbin and salinin 10 μg), 582 mg dextrin/malto-dextrin or other carrier (e.g., di-calcium phosphate or any other pharmaceutical grade carrier); and 18 mg aerated or fumed silica.


A free-flowing powder was prepared and encapsulated in suitable size of hard gelatin or vegetarian capsule available in the market. This formulation can be administered 3-4 times a day to get a therapeutic dose of 150-300 mg Neem leaf CO2 extract.


Formulation 3.


SCO2 Azadirachta indica leaf extract: 50 mg (with minimum 0.15 mg nimbolide; 6.5 μg nimbin and salinin 10 μg), 582 mg of water extract obtained from Neem leaf and 18 mg aerated or fumed silica.


A free-flowing powder was prepared and encapsulated in suitable size of hard gelatin or vegetarian capsule available in the market. This formulation can be administered 3-4 times a day to get a therapeutic dose of 150-300 mg Neem leaf CO2 extract.


Formulation 4.


Mouthwash formulation containing SCO2 Azadirachta indica leaf extract: 2.28 g (with minimum nimbolide: 7.2 mg; nimbin: 296.4 μg and salinin 456 μg), Peppermint (Mentha piperita) oil: 13.81 g, Spearmint (Mentha spicata) oil: 9.26 g, Clove Bud (Syzigium aromaticum) CO2 oil: 3.98 g, Tween 80: 20.68 g. 1.25 g of the said blend was diluted in 98.75 g in Base. The base contains water: 73.5 g, Aloe Vera water (200×): 10 g, sorbitol: 10 g, glycerin: 5.9 g, ascorbic acid: 0.5 g, potassium porbate: 0.1 g.


A liquid formulation was prepared. The same can be administered 3 times a day, 20 ml each time, to get a therapeutic dose of 15 mg Neem Leaf CO2 extract as described herein. The same can be administered 3 times a day, 20 ml each time, to get a therapeutic dose of 150 mg Neem Leaf CO2 extract as described herein.


Alternatively, a liposomal water based formulation was prepared using 2 gm of SCO2 extract as above with 95.6 gm of demineralized water with 2 gm of peppermint oil and 0.2 gm of Rosemary CO2 extract to obtain 100 gm of mouthwash formulation with minimum additives with standard pharmaceutical grade emulsifier like “Polysorbate 80” 0.2 gm using a high speed homogenizer.


Example 7: Cancer Preventive Effect of a Super Critical CO2 Neem Extract of Leaf (SCNE)

Cell Viability Assay:


Colorectal cancer cells, HCT116 and HT29 as well as normal rat colon cells IEC-6 cells, were plated in 96-well plates, next day the cells were serum starved for 24 hrs and treated with SCNE (0-75 μg/mL) and nimbolide (1-15 μM) for 48 h and 72 h. After treatment, cell viability was measured by MTT [3-(4, 5-Dimethylthiazol-2-yl)-2, 5-Diphenyltetrazolium Bromide] assay (Sigma Aldrich, MO) according to the manufacturer's instructions. Briefly, MTT (5 mg/mL) was added and plates were incubated at 37° C. for 4 h before dimethyl sulfoxide was added to each well. Finally, the absorbance of each well was read at a wavelength of 540 nm using a plate reader (Molecular Devices, Sunnyvale, Calif., USA). The results were expressed as a percentage of surviving cells over non-treated cells.


The results confirm that the SCNE is non-toxic to normal rat colon cells IEC-6 cells even at higher concentration of 50 μg/mL after 48 hrs (FIG. 14). The SCNE treated colorectal cancer cells, viz., HCT116 and HT29 exhibits 62% (FIG. 15) and 44% cell viability (FIG. 16) respectively at a concentration of 15 μg/mL at the end of 72 hrs and exhibits zero cell viability at a concentration of 40 μg/mL (FIG. 15) and 75 μg/mL (FIG. 16) at the end of 72 hrs. The nimbolide treated colorectal cancer cells, viz., HCT116 (FIG. 17) and HT29 (FIG. 18) exhibits 80% and 75% cell viability respectively at a concentration of 15 μg/mL at the end of 48 hrs.


The experiment described herein conclusively confirmed that the super critical CO2 neem leaf extract (SCNE extract) comprising a combination of nimbolide, nimbin and salinin possess higher therapeutic efficacy than the nimbolide alone.


Overall, the data suggests that SCNE effectively suppress the growth of human colorectal cancer through induction of apoptosis via pro-inflammatory pathway and NF-kB inhibition.

Claims
  • 1. A method of treating cancer in a subject, the method comprising: (a) identifying a subject in need of treatment; and(b) administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 2. The method of claim 1, further comprising a pharmaceutically acceptable excipient.
  • 3. The method of claim 1, wherein the subject is a human.
  • 4. The method of claim 1, wherein the pharmaceutically acceptable excipient is selected from the group di-calcium phosphate, distilled water, saline, aqueous glucose solution, alcohol (e.g. ethanol), surfactants, propylene glycol, tween-80 and polyethylene glycol; and oily carriers such as various animal and vegetable oils, white soft paraffin, paraffin, wax, glucose, fructose, sucrose, maltose, yellow dextrin, malt dextrin, white dextrin, aerosol, microcrystalline cellulose, calcium stearate, magnesium stearate, sorbitol, stevioside, corn syrup, lactose, citric acid, tartaric acid, malic acid, succinic acid, lactic acid, L-ascorbic acid, dl-alpha-tocopherol, glycerin, propylene glycol, glycerin fatty ester, poly glycerin fatty ester, sucrose fatty ester, sorbitan fatty ester, propylene glycol fatty ester, acacia, carrageenan, casein, gelatin, pectin, agar, vitamin B group, nicotinamide, calcium pantothenate, amino acids, aerated or fumed silica, calcium salts, pigments, flavors and preservatives.
  • 5. The method of claim 1, wherein the SCNE is administered in a dosage ranging from 50 mg to 1000 mg/day.
  • 6. The method of claim 5, wherein the amount of SCNE is about 50 mg to 1000 mg/day.
  • 7. The method of claim 1, wherein the amount of the nimbolide present in the composition is at least 3 mg/g, the amount of the nimbin present in the composition is at least 130 μg/g nimbin; and the amount of the salinin is at least 200 μg/g.
  • 8. The method of claim 1, wherein the SCNE comprises one or more liminoids.
  • 9. The method of claim 1, wherein the composition further comprises one or more tocopherols; and sesame oil.
  • 10. The method of claim 9, wherein the one or more tocopherols are alpha-tocopheraol, gamma-tocopherol, vitamin E or Rosemarinus officinalis.
  • 11. The method of claim 1, wherein the composition further comprises one or more tocopherols; sesame oil; and aerated or fumed silica.
  • 12. The method of claim 1, wherein the composition is in a form comprising a capsule.
  • 13. The method of claim 1, wherein the composition is administered orally.
  • 14. The method of claim 12, wherein the capsule is administered orally two or three times a day.
  • 15. The method of claim 1, wherein the cancer is a primary or secondary tumor.
  • 16. The method of claim 1, wherein the cancer is oral cancer or colon cancer.
  • 17. A method of reducing at least one inflammatory cytokine in serum of a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 18. The method of claim 17, wherein the amount of SCNE is about 50 mg to 75 mg.
  • 19. The method of claim 17, wherein the SCNE is administered in a dosage ranging from 50 mg to 1000 mg/day.
  • 20. The method of claim 17, wherein the amount of the nimbolide present in the composition is at least 3 mg/g; the amount of the nimbin present in the composition is at least 130 ng/g; and the amount of the salinin is at least 200 ng/g.
  • 21. The method of claim 17, wherein the SCNE comprises one or more liminoids.
  • 22. The method of claim 17, wherein the composition further comprises one or more tocopherols; and sesame oil.
  • 23. The method of claim 22, wherein the one or more tocopherols are alpha-tocopheraol, gamma-tocopherol, vitamin E or Rosemarinus officinalis.
  • 24. The method of claim 17, wherein the composition further comprises one or more tocopherols; sesame oil; and aerated or fumed silica.
  • 25. The method of claim 17, wherein the composition is in a form comprising a capsule.
  • 26. The method of claim 17, wherein the composition is administered orally.
  • 27. The method of claim 25, wherein the capsule is administered orally two or three times a day.
  • 28. The method of claim 17, wherein the composition further comprises a pharmaceutically acceptable excipient.
  • 29. The method of claim 17, wherein the subject is human.
  • 30. The method of claim 17, wherein the subject has been diagnosed with oral cancer or colon cancer prior to the administering step.
  • 31. The method of claim 17, wherein the at least one inflammatory cytokine is IFN-γ, IFN-β, TNF-α, IL-6 or IL-1.
  • 32. The method of claim 31, wherein the at least one inflammatory cytokine is IL-6 or TNF-α.
  • 33. The method claim 17, wherein the subject's serum has increased levels of at least one inflammatory cytokine when compared to a reference sample before the administration of the composition comprising therapeutically effective amount of a supercritical CO2 neem extract.
  • 34. The method of claim 17, further comprising determining the level of at least one inflammatory cytokine in one or more cells of a subject before the administration of the composition comprising a therapeutically effective amount of a supercritical CO2 neem extract, wherein the level of at least one inflammatory cytokine is higher when compared to a reference sample.
  • 35. A method of reducing inflammation in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 36. The method of claim 35, wherein the subject is human.
  • 37. The method of claim 35, wherein the subject has been diagnosed with oral cancer or colon cancer prior to the administering step.
  • 38. The method of claim 35, wherein the inflammation is reduced by decreasing the expression of one or more of IFN-γ, IFN-β, TNF-α, IL-6, IL-1, NF-κB, STAT3, COX1 or COX2.
  • 39. A method of treating a hyperproliferative disorder in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 40. The method of claim 39, further comprising a pharmaceutically acceptable excipient.
  • 41. The method of claim 39, wherein the subject is a human.
  • 42. The method of claim 39, wherein the subject has been diagnosed with a need for treatment the disorder prior to the administering step.
  • 43. The method of claim 39, wherein the hyperproliferative disorder is cancer.
  • 44. The method of claim 43, wherein the cancer is oral cancer or colon cancer.
  • 45. A method of suppressing expression of NFkB and cycloxygenase in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 46. The method of claim 45, further comprising a pharmaceutically acceptable excipient.
  • 47. The method of claim 45, wherein the subject has been diagnosed with a need for suppressing the expression of NFkB and cyclooxygenase prior to the administering step.
  • 48. The method of claim 45, wherein the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step.
  • 49. The method of claim 45, further comprising the step of identifying a subject in need of treatment of a disorder of uncontrolled cellular proliferation.
  • 50. The method of claim 49, wherein the disorder of uncontrolled cellular proliferation is a cancer.
  • 51. The method of claim 50, wherein the cancer is oral cancer.
  • 52. A method of suppressing expression of NFkB and cyclooxygenase in at least one cell, the method comprising the step of contacting at least one cell with an effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 53. The method of claim 52, further comprising a pharmaceutically acceptable excipient.
  • 54. The method of claim 52, wherein the at least one cell is a human cell.
  • 55. The method of claim 52, wherein the contacting is via administration to a subject.
  • 56. The method of claim 55, wherein the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step.
  • 57. A method of modifying epidermal growth factor receptor (EGFR) signaling activity in a subject, the method comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 58. The method of claim 57, further comprising a pharmaceutically acceptable excipient.
  • 59. The method of claim 57, wherein the subject is a human.
  • 60. The method of claim 57, wherein the subject has been diagnosed with a need for modifying EFGR signaling activity.
  • 61. The method of claim 57, wherein the modifying is inhibiting.
  • 62. The method of claim 57, wherein the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step.
  • 63. The method of claim 62, further comprising the step of identifying a subject in need of treatment of a disorder of uncontrolled cellular proliferation.
  • 64. The method of claim 63, wherein the disorder of uncontrolled cellular proliferation is oral cancer.
  • 65. A method of inducing apoptosis of a cell in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of a supercritical CO2 neem extract (SCNE), wherein the SCNE comprises nimbolide, nimbin and salinin.
  • 66. The method of claim 65, further comprising a pharmaceutically acceptable excipient.
  • 67. The method of claim 65, wherein the subject is a human.
  • 68. The method of claim 65, wherein the subject has been diagnosed with a need for treatment of a disorder of uncontrolled cellular proliferation prior to the administering step.
  • 69. The method of claim 68, further comprising the step of identifying a subject in need of treatment of a disorder of uncontrolled cellular proliferation.
  • 70. The method of claim 69, wherein the disorder of uncontrolled cellular proliferation is a cancer.
  • 71. The method of claim 70, wherein the cancer is colon cancer.
  • 72. A process for preparation of standardized CO2 extract of Azadirachta indica leaves comprising: a) powdering the clean and matured dried Azadirachta indica leaves having moisture content less than 12% to obtain powder with fine particles having size below 0.42 mm;b) subjecting the powder of step a) to supercritical CO2 extraction at a pressure varying between 80 Bar (80 kg/cm2) and 350 Bar (350 kg/cm2) at a temperature range of 31° C. to 45° C. at a flow rate of 10 to 40 kg of CO2 per kg of raw material;c) separating the CO2 extractives at a pressure varying between 40 Bar to 65 Bar at a temperature lower than the extraction temperature to obtain Extract A;d) subjecting the remaining residual powder after separating Extract ‘A’ to further extraction using mixture of CO2 and ethyl alcohol at the pressure ranging between 80 Bar to 350 Bar and at a temperature range of 31° C. to 45° C.;e) collecting the ethyl alcohol laced with extract from separator by reducing the solvent pressure between 40 Bar and 65 Bar at a temperature lower than the extraction temperature, followed by vacuum distillation of ethanol to obtain Extract B; andf) combining Extract A and Extract B to obtain standardized CO2 extract of Azadirachta indica leaves.
  • 73. The process as claimed in claim 72, wherein the extract A is optionally subjected to high velocity micro-jet or nozzle to get a particle size of 10 nm-100 nm.
  • 74. The process as claimed in claim 72, wherein the ethyl alcohol is used in an amount of 3 to 10% of the CO2.
  • 75. The process as claimed in claim 72, wherein, the separation temperature in step c) and collection temperature in step e) is maintained between 10° C. to 30° C.
  • 76. The process as claimed in claim 72, wherein, the vacuum distillation of ethanol is carried at temperature below 45° C.
  • 77. The process as claimed in claim 72, wherein, the standardized extract obtained in step f) comprises nimbolide in a minimum amount of 3 mg/gm; nimbin in a minimum amount of 130 μg/gm and salinin in a minimum amount of 200 μg/gm.
  • 78. A standardized CO2 extract of Azadirachta indica comprises nimbolide in a minimum amount of 3 mg/gm; nimbin in a minimum amount of 130 μg/gm and salinin in a minimum amount of 200 μg/gm.
  • 79. The standardized extract as claimed in claim 78, wherein, the extract further contains various other active phytoconstituents such as desacetylnimbin, azadiradione, azdirone, nimbolin, and nimbinene in minor amounts.
  • 80. A therapeutic herbal composition comprising standardized CO2 extract of Azadirachta indica as claimed in claim 78 or claim 79 in an effective amount of 50 to 300 mg along with one or more pharmaceutical carriers/excipients.
  • 81. The herbal composition as claimed in claim 80, wherein the extract comprises nimbolide in a minimum amount of 3 mg/gm; nimbin in a minimum amount of 130 μg/gm and salinin in a minimum amount of 200 μg/gm along with other active phytoconstituents, desacetylnimbin, azadiradione, azdirone, nimbolin, and nimbinene in minor amounts.
  • 82. The herbal composition as claimed in claim 80, wherein the pharmaceutical excipients/carriers are selected from the group consisting of distilled water, saline, aqueous glucose solution, alcohol (e.g., ethanol), surfactants, propylene glycol, tween-80 and polyethylene glycol; and oily carriers such as various animal and vegetable oils, white soft paraffin, paraffin, wax, glucose, fructose, sucrose, maltose, yellow dextrin, malt dextrin, white dextrin, aerosol, microcrystalline cellulose, calcium stearate, magnesium stearate, sorbitol, stevioside, corn syrup, lactose, citric acid, tartaric acid, malic acid, succinic acid, lactic acid, L-ascorbic acid, dl-alpha-tocopherol, glycerin, propylene glycol, glycerin fatty ester, poly glycerin fatty ester, sucrose fatty ester, sorbitan fatty ester, propylene glycol fatty ester, acacia, carrageenan, casein, gelatin, pectin, agar, vitamin B group, nicotinamide, calcium pantothenate, amino acids, aerated or fumed silica, calcium salts, pigments, flavors and preservatives.
  • 83. The herbal composition as claimed in claim 80, wherein the composition can be formulated into oral solid or liquid dosage forms.
Priority Claims (1)
Number Date Country Kind
201821021206 Sep 2019 IN national
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of the filing date of U.S. Provisional Application No. 62/725,484 which was filed on Aug. 31, 2018, and Indian Application No. 201821021206 which was filed on Sep. 6, 2019. The content of these earlier filed applications are hereby incorporated herein by reference in their entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/049059 8/30/2019 WO 00
Provisional Applications (1)
Number Date Country
62725484 Aug 2018 US