CN116710088A - JAK1 pathway inhibitors for the treatment of vitiligo - Google Patents

JAK1 pathway inhibitors for the treatment of vitiligo Download PDF

Info

Publication number
CN116710088A
CN116710088A CN202180082167.5A CN202180082167A CN116710088A CN 116710088 A CN116710088 A CN 116710088A CN 202180082167 A CN202180082167 A CN 202180082167A CN 116710088 A CN116710088 A CN 116710088A
Authority
CN
China
Prior art keywords
jak1
vitiligo
compound
subject
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180082167.5A
Other languages
Chinese (zh)
Inventor
P·史密斯
K·A·布朗
M·D·豪威尔
F·郭
J·李
桑托斯 L·L·多斯
B·伦伯格
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Incyte Corp
Original Assignee
Incyte Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Corp filed Critical Incyte Corp
Priority claimed from PCT/US2021/062419 external-priority patent/WO2022125670A1/en
Publication of CN116710088A publication Critical patent/CN116710088A/en
Pending legal-status Critical Current

Links

Abstract

The present disclosure relates to JAK1 pathway inhibitors and their use for the treatment of vitiligo.

Description

JAK1 pathway inhibitors for the treatment of vitiligo
Priority statement
The present application claims the benefit of priority from U.S. provisional application Ser. No. 63/122,574 filed on 8 th 12 and U.S. provisional application Ser. No. 63/246,688 filed on 21 9 and 2021, each of which is incorporated herein by reference in its entirety.
Technical Field
The present disclosure relates to JAK1 pathway inhibitors and their use for the treatment of vitiligo.
Background
Vitiligo is a chronic skin condition characterized by depigmenting plaques of the skin due to autoimmune destruction of melanocytes, and is estimated to affect 0.5-2.0% of the global population (regional). Vitiligo prevalence is similar in men and women, and there is no known difference in performance based on skin type or race. The average age of onset is 20 years old, but can occur at any age. The condition tends to progress over time. 15-25% of vitiligo patients also tend to suffer from at least one other autoimmune disorder. Non-segmental vitiligo is the most common subtype (up to 90% of vitiligo patients) and is characterized by symmetrical bilateral white spots. White spots are commonly found in the hands, feet, and perioral/facial areas.
Due to psychological and social factors, there is a potentially significant impact on the quality of life of patients suffering from vitiligo. The long-term nature and lack of effective treatment of vitiligo can have negative psycho-social effects on patients, affecting quality of life as with other skin diseases. Cosmetic sensitive areas such as the face and hands are affected with a significant impact on self-esteem and ultimately with psychological burden and quality of life; individuals with body surface areas exceeding 25% may have difficulty performing everyday tasks such as gardening, shopping or clothing selection, and social difficulties such as engaging in athletic activities and initiating and maintaining romantic relationships.
Clinical management aims at preventing pigment loss and inducing pigment regeneration. No drug is currently approved for the treatment of vitiligo patients. In general, first line therapy consists of topical corticosteroids and calcineurin inhibitors as used in the specification. Two-line therapy consists of phototherapy (narrowband UVB and psoralen and UVA) and systemic corticosteroids. Less common therapies include surgical grafting techniques and decolorizing treatments. Therefore, there is a need to develop new therapies for the treatment of vitiligo. The present application addresses this need and other needs.
Drawings
FIG. 1 is a graph depicting melanocyte proliferation,% confluence, and time (hours).
FIG. 2 is a graph depicting the effect of Compound 1 on a JAK 1-dependent cytokine using T cell compartments, protein T cell medium (pg/ml) and a specific JAK 1-dependent cytokine.
FIGS. 3A-3C are graphs depicting the effect of Compound 1 on growth factors and chemokines using T cell compartments, protein T cell medium (pg/ml) and IL-8 (A), HGF (B) and LIF (C).
FIG. 4 is a graph depicting the effect of Compound 1 on a JAK 1-dependent cytokine using melanocytes, protein melanocyte medium (pg/ml), and a specific JAK 1-dependent cytokine.
FIGS. 5A-5C are graphs depicting the effect of Compound 1 on growth factors and chemokines obtained using melanocytes, protein melanocyte medium (pg/ml) and IL-8 (A), HGF (B) and LIF (C).
Figure 6 depicts a summary of phase 2 randomized, double-blind, placebo-controlled dose range study of the efficacy and safety of compound 1.
Fig. 7 is a graph depicting melanocyte proliferation,% confluence, and time (hours).
FIG. 8 is a graph depicting the effect of ruxolitinib (ruxolitinib) on a JAK 1-dependent cytokine using T cell compartments, protein T cell medium (pg/ml) and a specific JAK 1-dependent cytokine.
FIGS. 9A-9B are graphs depicting the effect of ponatinib on growth factors and chemokines obtained using T cell compartments, protein T cell medium (pg/ml) and IL-8 (A) and LIF (B).
FIG. 10 is a graph depicting the effect of ponatinib on a JAK 1-dependent cytokine using melanocytes, protein melanocyte medium (pg/ml), and a specific JAK 1-dependent cytokine.
FIGS. 11A-11B are graphs depicting the effect of ponatinib on growth factors and chemokines obtained using melanocytes, protein melanocyte medium (pg/ml) and IL-8 (A) and LIF (B).
Disclosure of Invention
Provided herein are methods for treating vitiligo in a subject in need thereof, the methods comprising administering to the subject a therapeutically effective amount of a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof.
Provided herein is a JAK1 pathway inhibitor, or a pharmaceutically acceptable salt thereof, for use in treating vitiligo in a subject in need thereof.
Provided herein is the use of a JAK1 pathway inhibitor, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of vitiligo in a subject in need thereof.
Detailed Description
The invention provides, inter alia, a method of treating vitiligo in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof. In some embodiments, the vitiligo is non-segmental vitiligo.
In some embodiments, the invention provides a method for treating vitiligo in a subject, the method comprising administering to the subject a therapeutically effective amount of a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is more selective for JAK1 than JAK2, JAK3, and Tyk2.
In some embodiments, the JAK1 pathway inhibitor is 4- [3- (cyanomethyl) -3- (3 ',5' -dimethyl-1 h,1'h-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2, 2-trifluoro-1-methylethyl ] benzamide or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 pathway inhibitor is 4- [3- (cyanomethyl) -3- (3 ',5' -dimethyl-1 h,1'h-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2, 2-trifluoro-1-methylethyl ] benzamide phosphate.
In some embodiments, the vitiligo is non-segmental vitiligo.
In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about 5mg to about 95mg based on free base.
In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered at a daily dose of about 15mg, about 45mg, 75mg, or about 90mg, based on free base.
In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in combination with an additional therapeutic agent.
In some embodiments, the additional therapeutic agent comprises a Janus kinase inhibitor.
In some embodiments, the Janus kinase inhibitor comprises pontinib or a pharmaceutically acceptable salt thereof.
In some embodiments, the administering comprises administering a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable carrier or excipient.
In some embodiments, the vitiligo is non-segmental vitiligo. Non-segmented vitiligo is associated with some form of symmetry in the location of depigmented plaques. The non-segmental vitiligo comprises generalized vitiligo, acrofacial vitiligo, mucosa type vitiligo and focal vitiligo. In some embodiments, the vitiligo is generalized vitiligo. In some embodiments, the vitiligo is idiopathic vitiligo. In some embodiments, the vitiligo is acromioclavicular vitiligo. In some embodiments, the vitiligo is mucosal vitiligo. In some embodiments, the vitiligo is focal vitiligo. Generalized vitiligo is the most common category affecting about 0.5% of the global population, with an average age of onset of about 24 years, and the incidence of both men and women is about the same. Although there is no ethnicity difference, the disease may be more pronounced for darker individuals and thus may cause emotional distress.
In some embodiments, the vitiligo is segmental vitiligo. Segmental vitiligo is different from non-segmental vitiligo in appearance, etiology and prevalence. Segmental vitiligo often affects the skin area associated with the dorsal root of the spinal cord and is usually unilateral. The spread speed of segmental vitiligo is much faster than that of non-segmental vitiligo, and the disease course of segmental vitiligo is more stable/static under the condition of no treatment and is irrelevant to autoimmune diseases.
In some embodiments, the efficacy of the treatment methods disclosed herein can be established based on percent change from different baseline measurements using various indicators. For example, total% of pigment deprived surface area (BSA) (including facial and non-facial areas) may be used. BSA can be assessed by Palmar method as follows: the approximate size of the entire palm surface of the subject (i.e., palm plus 5 fingers) should be considered 1% bsa, and the approximate size of the subject's thumb should be considered 0.1% bsa.
As used herein, the term "face" may be generally defined to include the forehead to original hairline, cheek to mandible perpendicular to mandible, and area laterally from the mouth angle to the tragus. The "facial" area does not include the surface area of the lips, scalp, ears or neck, but includes the nose and eyelids.
In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in total depigmented surface area (T-BSA) of a subject of about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95%.
In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction of about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% in depigmented facial body surface area (F-BSA) of a subject.
In some embodiments, the area affected by vitiligo-induced depigmentation can be assessed using a Vitiligo Area Scoring Index (VASI). VASI is a comprehensive estimate based on the percentage of vitiligo affected (% BSA, using the Palmar method) and the extent of vitiligo focal pigment loss. In some embodiments, the degree of pigment loss may be determined and estimated to be the closest of the following percentages: 0%, 10%, 25%, 50%, 75%, 90% or 100%. When 100% of pigments are removed, pigments are not present; at 90%, pigment spots were present; at 75%, the pigment deprived area exceeds the colored area; at 50%, the pigment removal area and the coloring area are equal; at 25%, the colored area exceeds the pigment deprived area; at 10%, only depigmented spots were present. At 0% depigmentation, there is no depigmentation.
In some embodiments, a facial vitiligo area scoring index (F-VASI) is then obtained by multiplying the percentage of facial vitiligo involvement (% F-BSA) by the degree of pigment loss within the vitiligo lesions, and summing the values of all lesions.
In some embodiments, efficacy may be measured using systemic VASI (T-VASI) by multiplying the percentage of systemic vitiligo involvement (%t-BSA) by the extent of pigment loss within vitiligo lesions of all body parts and summing the values of all parts (range of possibilities, 0 to 100). For the purpose of T-VASI assessment, the body is divided into the following 6 independent and mutually exclusive sites: (1) head/neck (including scalp), (2) hands, (3) upper limbs (including armpits and excluding hands), (4) trunk (including genitals and excluding buttocks), (5) lower limbs (including buttocks and excluding feet), and (6) feet.
In some embodiments, efficacy may be assessed based on the percent change in T-VASI from baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a decrease in T-VASI in a subject. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in T-VASI in a subject of about 50% or greater. In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in a reduction in T-VASI in a subject of about 75% or greater. In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in a reduction in T-VASI in a subject of about 90% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in T-VASI in a subject of about 40% to about 90% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in T-VASI in a subject of about 50% to about 95% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in T-VASI in a subject of about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95%.
In some embodiments, efficacy may be assessed based on the proportion of subjects who achieve a reduction in T-VASI of about 50% or greater over a period of time. In some embodiments, efficacy may be assessed based on the proportion of subjects achieving a reduction in T-VASI of about 50% or greater, about 75% or greater, or about 90% or greater over a period of time.
In some embodiments, efficacy may be assessed based on the percent change of F-VASI from baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a decrease in F-VASI in a subject. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in F-VASI in a subject of about 50% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in F-VASI in a subject of about 75% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in F-VASI in a subject of about 90% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in F-VASI in a subject of about 40% to about 90% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in F-VASI in a subject of about 50% to about 95% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a reduction in F-VASI in a subject of about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95%.
Vitiligo degree score (VES) can be a validated measure representing the involvement of body mass vitiligo. VES uses pictures of 19 independent anatomical regions to assess the extent of vitiligo (%bsa).
Vitiligo degree score plus (VESplus) can be a validated metric representing the involvement of body mass vitiligo and a scale to evaluate perifollicular pigment regeneration patterns. VESplus uses pictures of 19 independent areas to assess the extent of vitiligo (% BSA) and their respective perifollicular pigment regeneration patterns, divided into 7 categories: 0%, 5%, 10%, 25%, 50%, 75% and 90%.
In some embodiments, VESplus may be used to evaluate efficacy based on the extent of vitiligo and improvement in perifollicular pigment regeneration patterns. Typically, VESplus uses BSA multiplied by a calculation of the extent of perifollicular pigment regeneration. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in VESplus in a subject. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a VESplus of about 50% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a VESplus of about 75% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a VESplus of about 90% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a VESplus of about 50% to about 90% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a VESplus of about 5% to about 75% or greater. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a VESplus of about 5% to about 90% or greater.
In some embodiments, efficacy may be assessed based on improvement in facial static investigator overall assessment (FSIGA) scores. The severity of facial vitiligo can be assessed using FSIGA with a 5-component scale (table 1).
TABLE 1
In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in FSIGA score of the subject as clear (0) or almost clear (1). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in FSIGA score of at least 2 points. In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in a subject's FSIGA score of clear (0) or nearly clear (1) and an improvement of at least 2 points.
In some embodiments, efficacy may be assessed based on improvement in static investigator overall assessment (SIGA) scores. The severity of vitiligo can be assessed using SIGA with a 5-component scale (table 2).
TABLE 2
In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's SIGA score of clearance (0) or near clearance (1). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in SIGA score of at least 2 points. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in SIGA score of at least 1, 2, or 3 points. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject having a SIGA score of clearance (0) or nearly clearance (1) and an improvement of at least 2 points.
Vitiligo is characterized by progressive loss of functional melanocytes with associated hearing abnormalities, including sensorineural hearing loss (SNHL) with prevalence ranging from 4.0% to 68.8% compared to healthy controls. During embryogenesis, precursor melanocytes (melanocytes) migrate to the dermis and also to the cochlea floor responsible for hearing high frequencies. It appears that inner ear melanocytes are important for cochlear hair cell function and normal hearing. The patient may perform a SNHL assessment at baseline to assess hearing and determine potential hearing improvement associated with JAK1 inhibitor (e.g., compound 1) exposure throughout the course of treatment. In some embodiments, efficacy may be assessed based on improvement in hearing of the subject. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's baseline hearing parameters.
In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in baseline hearing parameters of a subject of about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95%. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in baseline hearing parameters of a subject of about 50% or more.
In some embodiments, efficacy may be assessed based on improvement in the subject's response to the color matching problem, i.e., the Patient Reported Outcome (PRO). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to a color matching problem. For the color matching question, the facial image of the current subject (the subject may use a mirror) may be presented to the subject for reference and the subject may be asked to answer the following questions: "how does you treat to what extent does you match skin color between your treated facial vitiligo skin and normal facial skin? "possible responses are: (1) excellent, (2) excellent, (3) excellent, (4) poor, and (5) very poor. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in at least a 2 point improvement to the color matching problem. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in at least 1, 2, or 3 point improvement to the color matching problem.
In some embodiments, efficacy may be assessed based on improvement in the subject's estimated response PRO to a baseline facial photograph. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's assessment response to a baseline facial photograph. Vitiligo perceptibility scale (VNS) is a measure of patient reported success of vitiligo treatment, with a 5-component scale. The baseline facial photograph may be presented to the subject for reference, and the subject may be provided with a mirror to assess vitiligo on their face. The subject may be asked to answer the following questions: "how much more visible is vitiligo now compared with before treatment? "possible responses are: (1) more pronounced, (2) as pronounced, (3) slightly less pronounced, (4) less pronounced, and (5) no longer pronounced. How much is vitiligo now evident compared to before treatment? Response: (1) more pronounced, (2) as pronounced, (3) slightly less pronounced, (4) less pronounced, and (5) no longer pronounced. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a VNS score of 4 or 5.
In some embodiments, efficacy may be assessed based on improvement in response PRO of a subject to a change in overall impression of a facial patient-vitiligo (F-PaGIC-V). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to F-PaGIC-V. F-PaGIC-V is an assessment of improvement. It is a 7-component scale comparing the vitiligo area at baseline with the area of facial vitiligo treated in the subject. The baseline photograph and facial image of the current subject (which may provide a mirror for the subject) are presented to the subject for reference. The subject may be asked to answer the following questions: since you received treatment in this study, you received study medication to treat vitiligo on the face: (1) very much improvement, (2) much improvement, (3) little improvement, (4) no change, (5) little deterioration, (6) much deterioration, and (7) much deterioration. In some embodiments, compound 1 and/or methods of use described herein result in an F-PaGIC-V score of 1 or 2.
In some embodiments, efficacy may be assessed based on improvement of the subject's response PRO to a general patient's changing overall impression-vitiligo (F-PaGIC-V). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to T-PaGIC-V. T-PaGIC-V is an assessment of improvement. It is a 7-component scale comparing the vitiligo area at baseline with the whole body vitiligo area of the subject treated. The subject may be asked to answer the following questions: since you received treatment in this study, you received systemic vitiligo after study drug treatment: (1) very much improvement, (2) much improvement, (3) little improvement, (4) no change, (5) little deterioration, (6) much deterioration, and (7) much deterioration. In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in a T-PaGIC-V score of 1 or 2.
In some embodiments, efficacy may be assessed based on improved PRO of a subject's self-assessed vitiligo degree score (SA-VES). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to SA-VES over baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to SA-VES that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. The self-assessed vitiligo degree score (SA-VES) can be a validated tool that allows the patient to select the degree of disease (via image presentation) in 12 anatomical regions.
In some embodiments, efficacy may be assessed based on improved PRO of vitiligo quality of life (vitqol) in a subject. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to VitiQoL relative to baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to VitiQoL that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. VitiQoL is a 15-item quality of life assessment that requires a subject to score various aspects of his condition over the past month using a 7-point scale ("no" to "always").
In some embodiments, efficacy may be assessed based on improved PRO of the dermatological quality of life index (DLQI) of the subject. In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in an improvement in a subject's response to DLQI relative to baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to DLQI that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. DLQI is a validated questionnaire of 10 questions used to measure the extent of the effect of skin problems on a subject over the past 7 days. The subject may answer the questionnaire with (1) very many, (2) many, (3) a few, or (4) no at all. The questionnaire was analyzed under the following 6 titles: symptoms and sensations (questions 1 and 2); daily activities (questions 3 and 4); leisure (questions 5 and 6); work and school (problem 7); personal relationships (questions 8 and 9); and treatment (problem 10).
In some embodiments, efficacy may be assessed based on improved PRO in the subject's Hospital Anxiety and Depression Scale (HADS). In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to HADS relative to baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to HADS that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. HADS is a 14-item questionnaire that is used to assess the level of anxiety and depression an individual is currently experiencing. There were 7 questions each for measuring anxiety and for measuring depression, with 4 possible responses (response scores of 0, 1, 2 or 3) for each question. Scores for anxiety and depression were calculated separately.
In some embodiments, efficacy may be assessed based on improved PRO of WHO-5 in the subject. In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in an improvement in a subject's response to WHO-5 relative to baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to WHO-5 that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. WHO-5 may be a validated, self-administered 5-item questionnaire designed to evaluate mental health over the past 2 weeks, useful as a measure of the outcome of both desired and undesired therapeutic effects. The questionnaire consisted of 5 statements, which were scored by the interviewee according to the following scale: 0 = none; 1 = sometimes; 2 = less than half the time; 3 = more than half the time; 4 = most of the time; 5=always.
The raw score is calculated by adding the numbers of 5 answers ranging from 0 to 25, where 0 represents the worst possible quality of life and 25 represents the best possible quality of life. Scores below 13 indicate poor health.
In some embodiments, efficacy may be assessed based on improvement of a subject's medication satisfaction questionnaire (TSQM). In some embodiments, a JAK1 inhibitor (e.g., compound 1) and/or method of use described herein results in an improvement in a subject's response to TSQM relative to baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to TSQM that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. The TSQM may be a validated 9-item questionnaire that measures the satisfaction of a subject with a treatment using a recall period of the last 2 to 3 weeks or since the last treatment use. The questionnaire uses a 7-point scale for each question.
In some embodiments, efficacy may be assessed based on improvement of the subject's EQ-5D-5L questionnaire. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in an improvement in a subject's response to an EQ-5D-5L questionnaire relative to baseline. In some embodiments, JAK1 inhibitors (e.g., compound 1) and/or methods of use described herein result in a subject's response to an EQ-5D-5L questionnaire that is improved by about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% relative to baseline. The EQ-5D-5L questionnaire is a standardized validated tool that can be used as a measure of health outcome. The EQ-5D-5L questionnaire will provide data for economic models and analysis, including developing health utility or Quality Adjusted Life Years (QALY). The EQ-5D-5L questionnaire consists of the following 2 parts: EQ-5D describes a system and EQ VAS. The description system includes five aspects: locomotor ability, self-care, daily activities, pain/discomfort and anxiety/depression.
There are 5 classes per aspect: grade 1 is "no problem", grade 2 is "slight problem", grade 3 is "moderate problem", grade 4 is "severe problem", and grade 5 is "extreme problem". This portion of the EQ-5D-5L questionnaire provides a descriptive profile that can be used to generate a health status profile. For example, a subject in "health state 12345" has no problem in activity ability, has a slight problem in self-care (washing or dressing), has a moderate problem in daily activities, has severe pain or discomfort, and is extremely anxious or depressed. Each health status may be assigned a summary index score based on the social preference weights of the health status. These weights are sometimes referred to as utilities and are typically used to calculate QALY for health economic analysis. The health status index score typically ranges from less than 0 (where 0 is a health status value corresponding to death; negative values represent worse than death) to 1 (fully healthy values), with higher scores representing higher health utility. Health status preferences generally represent country or region values and thus may vary from country to country/region. EQ VAS records the subject's self-assessed health status on a vertical visual analog scale (0 to 100), with endpoints marked as "best health status you can imagine" (100 points) and "worst health status you can imagine" (0 points).
The methods described herein utilize JAK1 pathway inhibitors, particularly JAK1 selective inhibitors. JAK1 selective inhibitors are compounds that preferentially inhibit JAK1 activity over other Janus kinases. JAK1 plays a central role in many cytokine and growth factor signaling pathways that, when deregulated, can lead to or contribute to disease states. For example, IL-6 (pro-inflammatory cytokine) levels are elevated in patients with vitiligo (Singh et al, indian JDermatology,2012, 1 month-2 months; 57 (1): 12-14). In other autoimmune diseases and cancers, elevated systemic levels of inflammatory cytokines that activate JAK1 may also lead to disease and/or related symptoms. Thus, patients with autoimmune diseases of vitiligo may benefit from JAK1 inhibition. Selective inhibitors of JAK1 may be effective while avoiding unnecessary and potentially adverse effects of inhibiting other JAK kinases.
In some embodiments, a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof is more selective for JAK1 than JAK2, JAK3, and TYK2 (i.e., a JAK1 selective inhibitor). For example, a compound described herein, or a pharmaceutically acceptable salt thereof, preferentially inhibits JAK1 over one or more of JAK2, JAK3, and TYK 2. In some embodiments, the compound preferentially inhibits JAK1 relative to JAK2 (e.g., has JAK2/JAK1 IC 50 Ratio of>1). In some embodiments, the compound or salt is about 10-fold more selective for JAK1 relative to JAK 2. In some embodiments, such as by measuring IC at 1mM ATP 50 The compound or salt is calculated to be about 3-fold, about 5-fold, about 10-fold, about 15-fold, or about 20-fold more selective for JAK1 relative to JAK2 (see, e.g., example a).
In some embodiments, the JAK1 pathway inhibitor is a compound of table 3 or a pharmaceutically acceptable salt thereof. The compounds in table 3 are selective JAK1 inhibitors (relative to selectivity for JAK2, JAK3 and TYK 2). IC obtained by the method of example A at 1mM ATP 50 The values are shown in Table 3Is a kind of medium.
The compounds of table 3 can be prepared by synthetic procedures described, for example, in the following documents: U.S. patent publication No. 2011/0224190 submitted at 3/9/2011, U.S. patent publication No. 2014/0343030 submitted at 5/16/2014, U.S. patent publication No. 2014/01231198 submitted at 10/31/2013, U.S. patent publication No. 2010/0298334 submitted at 21/2010, U.S. patent publication No. 2011/0059951 submitted at 8/31/2011, U.S. patent publication No. 2012/0149681 submitted at 11/18/2011, U.S. patent publication No. 2012/0149682 submitted at 11/18/2012, U.S. patent publication No. 2013/0018034 submitted at 6/19/2012/2013/0045963 submitted at 8/17 and U.S. patent publication No. 2014/0005166 submitted at 5/17/2013, each of which is incorporated herein by reference in its entirety.
TABLE 3 Table 3
/>
/>
/>
/>
/>
/>
/>
/>
/>
+ means <10nM (for assay conditions, see example A)
++ means +.ltoreq.100 nM (for assay conditions, see example A)
++ is less than or equal to 300nM in the case of the conditions of the measurement, see example A)
a Data for enantiomer 1
b Data for enantiomer 2
In some embodiments, the JAK1 pathway inhibitor is 4- [3- (cyanomethyl) -3- (3 ',5' -dimethyl-1 h,1'h-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2, 2-trifluoro-1-methylethyl ] benzamide (compound 1) or a pharmaceutically acceptable salt thereof. In some embodiments, the JAK1 pathway inhibitor is 4- [3- (cyanomethyl) -3- (3 ',5' -dimethyl-1 h,1'h-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2, 2-trifluoro-1-methylethyl ] benzamide phosphate. Compound 1 and salts thereof can be prepared by the procedure described in US 9,382,231 (see e.g., example 7) filed on 5 months 16 of 2014, which is incorporated herein by reference in its entirety.
In some embodiments, the JAK1 pathway inhibitor is selected from the group consisting of compounds described in the following documents: U.S. patent publication No. 2011/0224190 submitted at 3/9/2011, U.S. patent publication No. 2014/0343030 submitted at 5/16/2014, U.S. patent publication No. 2014/01231198 submitted at 10/31/2013, U.S. patent publication No. 2010/0298334 submitted at 21/2010, U.S. patent publication No. 2011/0059951 submitted at 8/31/2011, U.S. patent publication No. 2012/0149681 submitted at 11/18/2011, U.S. patent publication No. 2012/0149682 submitted at 11/18/2012, U.S. patent publication No. 2013/0018034 submitted at 6/19/2012/2013/0045963 submitted at 8/17 and U.S. patent publication No. 2014/0005166 submitted at 5/17/2013, each of which is incorporated herein by reference in its entirety.
In some embodiments, the JAK1 pathway inhibitor is a compound of formula I
Or a pharmaceutically acceptable salt thereof, wherein:
x is N or CH;
l is C (=o) or C (=o) NH;
a is phenyl, pyridinyl or pyrimidinyl, each of which is optionally substituted with 1 or 2 independently selected R 1 Group substitution; and is also provided with
Each R 1 Independently fluorine or trifluoromethyl.
In some embodiments, the compound of formula I is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinoyl ] piperidin-4-yl } -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of formula I is 4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N- [ 4-fluoro-2- (trifluoromethyl) phenyl ] piperidine-1-carboxamide, or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound of formula I is [3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -1- (1- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperidin-4-yl) azetidin-3-yl ] acetonitrile or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 pathway inhibitor is a compound of formula II
Or a pharmaceutically acceptable salt thereof, wherein:
R 2 is C 1-6 Alkyl, C 1-6 Haloalkyl, C 3-6 Cycloalkyl or C 3-6 cycloalkyl-C 1-3 Alkyl, wherein the C 1-6 Alkyl, C 3-6 Cycloalkyl and C 3-6 cycloalkyl-C 1-3 Alkyl groups are each optionally substituted with 1, 2 or 3 groups independently selected from fluorine, -CF 3 And methyl;
R 3 is H or methyl;
R 4 h, F or Cl;
R 5 is H or F;
R 6 is H or F;
R 7 is H or F;
R 8 is H or methyl;
R 9 is H or methyl;
R 10 is H or methyl; and is also provided with
R 11 Is H or methyl.
In some embodiments, the JAK1 pathway inhibitor is a compound of formula III
Or a pharmaceutically acceptable salt thereof, wherein:
Cy 4 is a tetrahydro-2H-pyran ring optionally substituted with 1 or 2 groups independently selected from: CN, OH, F, cl, C 1-3 Alkyl, C 1-3 Haloalkyl, cyano-C 1-3 Alkyl, HO-C 1-3 Alkyl groupAmino, C 1-3 Alkylamino and di (C) 1-3 Alkyl) amino, wherein said C 1-3 Alkyl and di (C) 1-3 Alkyl) amino groups optionally being selected from F, cl, C, 1, 2 or 3 independently 1-3 Alkylaminosulfonyl and C 1-3 A substituent of alkylsulfonyl; and is also provided with
R 12 is-CH 2 -OH、-CH(CH 3 ) -OH or-CH 2 -NHSO 2 CH 3
In some embodiments, the compound of formula III is ((2R, 5 s) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about 10mg to about 100mg based on free base. Thus, in some embodiments, the selective JAK1 pathway inhibitor is administered in a daily dose of about 10mg, about 15mg, about 20mg, about 25mg, about 30mg, about 35mg, about 40mg, about 45mg, about 50mg, about 55mg, about 60mg, about 65mg, about 70mg, about 75mg, about 80mg, about 85mg, about 90mg, about 95mg, or about 100mg based on free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about 1mg to about 100mg based on free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about 10mg to about 80mg based on free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered at a daily dose of about 90mg based on free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered at a daily dose of about 75mg based on free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered at a daily dose of about 45mg based on free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered at a daily dose of about 15mg based on free base.
The term "about" refers to "about" (e.g., about 10% of the indicated value is added or subtracted).
In some embodiments, the JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof is administered as one or more sustained release dosage forms, each comprising a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof.
In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered orally.
The embodiments described herein are intended to be combined in any suitable combination as if the embodiments were the multiple dependent claims (e.g., embodiments related to selective JAK1 pathway inhibitors and dosages thereof, embodiments related to any salt form of the compounds disclosed herein, embodiments related to each type of cytokine-related disease or disorder, and embodiments related to compositions and/or administration may be combined in any combination).
For the sake of brevity only, all possible combinations are not individually listed herein.
The compounds described herein may be asymmetric (e.g., have one or more stereocenters). Unless otherwise indicated, all stereoisomers such as enantiomers and diastereomers are contemplated. Compounds containing asymmetrically substituted carbon atoms may be isolated in optically active or racemic forms. Methods of how to prepare optically active forms from non-optically active starting materials are known in the art, for example by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, c=n double bonds, etc., may also be present in the compounds described herein, and all such stable isomers are contemplated by the present invention. Cis and trans geometric isomers of the compounds of the present disclosure are described and may be separated as mixtures of isomers or individual isomeric forms.
In some embodiments, the compound has an (R) -configuration. In some embodiments, the compound has an (S) -configuration.
Resolution of the racemic mixture of the compounds can be carried out by any of a variety of methods known in the art. An exemplary method includes fractional recrystallization using a chiral resolving acid, which is an optically active salified organic acid. Suitable resolving agents for use in the fractional recrystallisation process are for example optically active acids, such as tartaric acid in D and L form, diacetyl tartaric acid, dibenzoyl tartaric acid, mandelic acid, malic acid, lactic acid or various optically active camphorsulphonic acids such as β -camphorsulphonic acid. Other resolving agents suitable for use in the fractional crystallization process include stereoisomerically pure forms (e.g., S and R forms, or diastereoisomerically pure forms) of alpha-methylbenzylamine, 2-phenylglycinol, norephedrine, ephedrine, N-methyl ephedrine, cyclohexylethylamine, 1, 2-diaminocyclohexane, and the like.
Resolution of the racemic mixture may also be carried out by eluting on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable eluting solvent compositions can be determined by those skilled in the art.
The compounds described herein also include tautomeric forms. Tautomeric forms result from the exchange of single bonds with adjacent double bonds and concomitant proton migration. Tautomeric forms include proton transfer tautomers, which are isomerically protonated states of the same empirical formula and total charge. Examples of proton transfer tautomers include keto-enol pairs, amide-imide pairs, lactam-lactam pairs, enamine-imine pairs, and cyclic forms in which a proton may occupy two or more positions of the heterocyclic system, such as 1H-and 3H-imidazole, 1H-, 2H-and 4H-1,2, 4-triazole, 1H-and 2H-isoindole, and 1H-and 2H-pyrazole. Tautomeric forms may be in equilibrium or sterically locked into one form by appropriate substitution.
The compounds described herein may also include isotopically-labeled compounds of the present disclosure. An "isotopically" or "radiolabeled" compound is a compound of the disclosure in which one or more atoms are replaced or substituted with an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated into the compounds of the present disclosure include, but are not limited to 2 H (also denoted by D, deuterium), 3 H (also denoted by T, deuterium), 11 C、 13 C、 14 C、 13 N、 15 N、 15 O、 17 O、 18 O、 18 F、 35 S、 36 Cl、 82 Br、 75 Br、 76 Br、 77 Br、 123 I、 124 I、 125 I and 131 I. for example, one or more hydrogen atoms in the compounds of the present disclosure may be replaced by deuterium atoms (e.g., formula (I), (II) or (III) or C of a compound of table 3) 1-6 One or more hydrogen atoms of the alkyl group may be optionally substituted with deuterium atoms, e.g. -CD 3 substituted-CH 3 ). As used herein, the term "compound" is intended to include all stereoisomers, geometric isomers, tautomers and isotopes of the depicted structures, unless the name indicates a particular stereoisomer. Unless otherwise indicated, a compound identified herein by name or structure as one particular tautomeric form is intended to include other tautomeric forms.
All compounds and pharmaceutically acceptable salts thereof may be present with other substances such as water and solvents (e.g., hydrates and solvates), or may be isolated.
In some embodiments, a compound described herein, or a salt thereof, is substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it is formed or detected. Partial separations may include, for example, compositions enriched in the compounds described herein. Substantial isolation may include compositions containing at least about 50 wt%, at least about 60 wt%, at least about 70 wt%, at least about 80 wt%, at least about 90 wt%, at least about 95 wt%, at least about 97 wt%, or at least about 99 wt% of a compound described herein, or a salt thereof. Methods for isolating compounds and salts thereof are routine in the art.
The phrase "pharmaceutically acceptable" is used herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As used herein, the expressions "ambient temperature" and "room temperature" or "rt" are understood in the art and generally refer to a temperature (e.g., reaction temperature) of about the room temperature at which the reaction is carried out, such as a temperature of about 20 ℃ to about 30 ℃.
The invention also includes pharmaceutically acceptable salts of the compounds disclosed herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting the acid or base moiety present into its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues (e.g., amines); basic or organic salts of acidic residues (e.g., carboxylic acids); etc. Pharmaceutically acceptable salts of the invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. Pharmaceutically acceptable salts of the invention can be synthesized from the parent compound containing a basic or acidic moiety by conventional chemical methods. Typically, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent or in a mixture of both; in general, non-aqueous media such as ether, ethyl acetate, alcohols (e.g., methanol, ethanol, isopropanol, or butanol) or Acetonitrile (ACN) are preferred. A list of suitable salts is found in Remington's Pharmaceutical Sciences, 17 th edition, mack Publishing Company, easton, pa.,1985, pages 1418 and Journal of Pharmaceutical Science,66,2 (1977), each of which is incorporated herein by reference in its entirety.
As used herein, the terms "subject," "individual," or "patient" are used interchangeably to refer to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses, or primates, and most preferably humans. In some embodiments, a "subject," "individual," or "patient" is in need of such treatment.
In some embodiments, the inhibitor is administered in a therapeutically effective amount. As used herein, the phrase "therapeutically effective amount" refers to the amount of an active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, subject, or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
As used herein, the term "treatment" refers to one or more of the following: (1) inhibiting a disease; for example, inhibiting a disease, disorder, or condition in an individual experiencing or exhibiting the pathology or symptom of the disease, disorder, or condition (i.e., preventing further development of the pathology and/or condition); (2) alleviation of disease; for example, alleviating a disease, disorder or condition (i.e., reversing pathology and/or symptoms) in an individual experiencing or exhibiting the pathology or symptoms of the disease, disorder or condition, e.g., reducing the severity of the disease.
In some embodiments, the JAK1 inhibitor may prevent vitiligo in an individual who may be susceptible to the disease. The term "preventing" refers to preventing the occurrence of a disease in a patient who may be susceptible to the disease but has not yet experienced or exhibited the pathology or symptomology of the disease.
Combination therapy
The methods described herein may further comprise administering one or more additional therapeutic agents. The one or more additional therapeutic agents may be administered to the patient simultaneously or sequentially. Methods other than compound 1 may be used (e.g., topically) to administer one or more additional therapeutic agents.
In some embodiments, the additional therapeutic agent is selected from other JAK inhibitors. Additional JAK inhibitors may include ATI-50002 (JAK 1/3 selectivity). Additional JAK inhibitors may include PF-06651600 (JAK 3 selective). Additional JAK inhibitors may include PF06700841 (JAK 1/TYK2 selectivity). Additional JAK inhibitors may include lapatinib (upadacrinib). Additional JAK inhibitors may include Abrocitinib (JAK 1 selectivity). Additional JAK inhibitors may include Cerdulatinib (JAK 1/SYK selectivity). Additional JAK inhibitors may include decarbavantinib (TYK 2 selectivity).
In some embodiments, the additional therapeutic agent is selected from antioxidants. The antioxidant is selected from pseudocatalase, vitamin E, vitamin C, ubiquinone, lipoic acid, white velvet keels (Polypodium leucotomos), catalase/superoxide dismutase combination, and Ginkgo (Ginkgo biloba). In some embodiments, the antioxidant may be further administered in combination with phototherapy. The administration of antioxidants during or prior to phototherapy aims at counteracting oxidative stress caused by the UV radiation itself, thereby improving the phototherapy effect.
In some embodiments, the additional therapeutic agent is a wingless-associated integration site (Wnt) agonist. Wnt agonists may include SKL2001.
In some embodiments, the additional therapeutic agent is a melanocortin 1 receptor (MC 1R) agonist.
In some embodiments, the additional therapeutic agent is an inflammatory mediator. Inflammatory mediators may include prostaglandin E2 (PGE) 2 ) And bimatoprost (a synthetic analog of prostaglandin F2 alpha).
In some embodiments, the additional therapeutic agent is an antimetabolite. The antimetabolite may include 5-fluorouracil.
In some embodiments, the additional therapeutic agent comprises plasmid HSP70i gene therapy. Plasmid HSP70i gene therapy may include HSP70iQ435A.
In some embodiments, the additional therapeutic agent is selected from the group consisting of a topical corticosteroid, an immunomodulator, a calcineurin inhibitor, and phototherapy. In some embodiments, the additional therapy is a systemic steroid or an immunosuppressant.
In some embodiments, the additional therapeutic agent comprises a steroid (e.g., an orally administered steroid), including a systemic steroid. Steroid treatment may include oral steroid micropulse therapy (e.g., using betamethasone and/or dexamethasone).
In some embodiments, the topical corticosteroid is selected from the group consisting of synergistic betamethasone dipropionate, beclomethasone propionate, diflorasone diacetate, halobetaxofenadine propionate (halobetasol propionate amcinonide), betamethasone valerate, dexamethasone, diflorasone diacetate, fluocinolone acetonide, halcinonide, and triamcinolone acetonide (triamcinolone acetonide).
In some embodiments, the additional therapeutic agent comprises an immunomodulatory agent. Immunomodulatory agents may include PDE4 inhibitors (e.g., apremilast (e.g., oral) or cliborole (e.g., topical)). Immunomodulatory agents may include anti-CD 20 therapies (e.g., ofatumumab). Immunomodulatory agents may include anti-CD 19 therapies (e.g., tafasitaumab). Immunomodulators may include anti-IL 15 therapies (e.g., AMG714 monoclonal antibodies). Immunomodulatory agents may include anti-IL 36 therapies (e.g., ismidolimab (imsidolimab) and se Bai Suoli mab (spisolimab)). Immunomodulatory agents may include anti-tnfα therapies such as etanercept (etanercept) and infliximab (infliximab). Immunomodulators may include anti-CD 122 therapies.
In some embodiments, the immunomodulator is selected from the group consisting of alprostadil, clenbuterol, afalanolide, rituximab, ofatuzumab, tafamuzumab, minocycline, latanoprost, zinc, tofacitinib, AMG 714 monoclonal antibody, iskedolizumab, span Bai Suoli monoclonal antibody cyclosporine, etanercept, infliximab, cyclophosphamide, cyclosporine, methotrexate, and oxo-dihydro-acridine sodium acetate (ODHAA).
In some embodiments, the calcineurin inhibitor is selected from tacrolimus (tacrolimus) (FK-506) and pimecrolimus (pimecrolimus).
In some embodiments, phototherapy includes exposure to ultraviolet light (e.g., excimer lamps or lasers).
In some embodiments, the additional therapeutic agent is a Janus kinase inhibitor. In some embodiments, the Janus kinase inhibitor is pontinib or a pharmaceutically acceptable salt thereof. In some embodiments, the rucotinib is administered topically. Topical administration of ponatinib is described in more detail in U.S. patent publication No. 2011/0288107 and Persaud et al, "Plasma pharmacokinetics and distribution of ruxolitinib into skin following oral and topical administration in minipigs" Int J Pharm,590:119889, 11/2020, 30/month PMID 32949620, filed 3/0288107, 2011, which is incorporated herein by reference in its entirety.
In some embodiments, the additional therapeutic agent is an IL-6 antagonist or a receptor antagonist. In some embodiments, the IL-6 receptor antagonist is tolizumab (Tocilizumab).
In some embodiments, the methods described herein may also include use in combination with surgical techniques. In some embodiments, the surgical technique comprises surgical skin grafting.
Pharmaceutical formulation and dosage form
When used as a medicament, JAK1 pathway inhibitors or pharmaceutically acceptable salts thereof may be administered in the form of a pharmaceutical composition. These compositions may be prepared in a manner well known in the pharmaceutical arts and may be administered by a variety of routes depending on whether local or systemic treatment is desired and the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and mucosal, including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal administration), oral or parenteral. Parenteral administration includes intravenous, intra-arterial, subcutaneous, intraperitoneal, intramuscular, or injection or infusion; or intracranial administration, such as intrathecal or intraventricular administration. Parenteral administration may be in the form of a single bolus dose or may be accomplished, for example, by a continuous infusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, foams, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily matrices, thickeners and the like may be necessary or desirable.
The present invention also includes pharmaceutical compositions comprising as an active ingredient a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers (excipients). In some embodiments, the composition is suitable for topical application. In preparing the compositions of the present invention, the active ingredient is typically admixed with, diluted by, or enclosed within an excipient, for example, in the form of a capsule, sachet, paper or other container. When the excipient acts as a diluent, it may be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient. Thus, the composition may take the form: tablets, pills, powders, troches, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing the formulation, the active compound may be milled to provide the appropriate particle size and then combined with the other ingredients. If the active compound is substantially insoluble, it may be milled to a particle size of less than 200 mesh. If the active compound is substantially soluble in water, the particle size may be adjusted by milling to provide a substantially uniform distribution in the formulation, for example about 40 mesh.
The JAK1 pathway inhibitor may be milled using known milling procedures such as wet milling to obtain a particle size suitable for tablet formation and other formulation types. Finely divided (nanoparticulate) formulations of JAK1 selective inhibitors can be prepared by methods known in the art, see for example international application No. WO 2002/000196.
The compositions may be formulated in unit dosage forms, each dosage containing an amount of the active ingredient in free form or in salt form. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
In the methods and uses of the invention, similar doses of the compounds described herein may be used.
The active compounds can be effective over a wide dosage range and are generally administered in pharmaceutically effective amounts. However, it will be appreciated that the amount of the compound actually administered is generally determined by a physician in the light of the relevant circumstances, including the disorder to be treated, the route of administration selected, the actual compound administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, and the like.
To prepare solid compositions, such as tablets, the primary active ingredient is admixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When these preformulated compositions are referred to as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing, for example, from about 0.1 to about 1000mg of the active ingredient of the present invention.
The tablets or pills of the invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, a tablet or pill may comprise an inner dosage component and an outer dosage component, the latter in the form of an envelope over the former. The two components may be separated by an enteric layer that serves to resist disintegration in the stomach and allows the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials may be used for such enteric layers or coatings, including a variety of polymeric acids and mixtures of polymeric acids with materials such as shellac, cetyl alcohol and cellulose acetate.
Liquid forms which may be incorporated into the compounds and compositions of the present invention for oral administration or administration by injection include aqueous solutions, suitably flavored syrups, aqueous or oily suspensions and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable aqueous or organic solvents or mixtures thereof, and powders. The liquid or solid composition may comprise suitable pharmaceutically acceptable excipients as described above. In some embodiments, the composition is administered by the oral or nasal respiratory route to produce a local or systemic effect. The composition may be atomized by using an inert gas. The nebulized solution may be drawn directly from the nebulizing device or the nebulizing device may be connected to a mask, tent, or intermittent positive pressure ventilator. The solution, suspension or powder composition may be administered orally or nasally from a device that delivers the formulation in a suitable manner.
The topical formulations may contain one or more conventional carriers. In some embodiments, the ointment may contain water and one or more hydrophobic carriers selected from the group consisting of: such as liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white vaseline, and the like. The carrier composition of the cream may be based on a combination of water with glycerol and one or more other components, such as glycerol monostearate, PEG-glycerol monostearate, and cetostearyl alcohol. The gels may be formulated using isopropanol and water, if appropriate in combination with other components (e.g., glycerol, hydroxyethylcellulose, etc.).
The amount of the compound or composition to be administered to a patient will vary depending on the drug being administered, the purpose of administration (e.g., prophylaxis or treatment), the condition of the patient, the mode of administration, and the like. In therapeutic applications, the composition may be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. The effective dosage will depend on the condition being treated and will be determined by the attending physician based on factors such as the severity of the disease, the age, weight and general condition of the patient.
The composition administered to the patient may be in the form of a pharmaceutical composition as described above. These compositions may be sterilized by conventional sterilization techniques or may be sterile filtered. The aqueous solution may be packaged for use as such or lyophilized, the lyophilized formulation being combined with a sterile aqueous carrier prior to administration. The pH of the compound formulation is typically between 3 and 11, more preferably 5 to 9 and most preferably 7 to 8. It will be appreciated that the use of certain of the foregoing excipients, carriers or stabilizers will result in the formation of pharmaceutically acceptable salts.
The therapeutic dosage of the compounds of the invention may vary depending upon, for example, the particular use for which the treatment is intended, the mode of administration of the compounds, the health and condition of the patient, and the discretion of the prescribing physician. The proportion or concentration of the compounds described herein in the pharmaceutical composition may vary depending on a number of factors including the dosage, chemical characteristics (e.g., hydrophobicity) and route of administration. The dosage may depend on, for example, the following variables: the type and extent of progression of the disease or disorder, the general health of the particular patient, the relative biological efficacy of the compound selected, the formulation of the excipients, and the route of administration thereof. The effective dose can be deduced from dose response curves derived from in vitro or animal model test systems.
The compositions of the invention may also include one or more additional pharmaceutical agents, such as chemotherapeutic agents, steroids, anti-inflammatory compounds, or immunosuppressants, examples of which are listed herein.
Medicine box
The invention also encompasses kits useful, for example, in the treatment and/or prevention of vitiligo comprising one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound described herein. Such kits may also include one or more different conventional kit components, such as containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as would be apparent to one of skill in the art, if desired. Instructions for the amount of the component to be administered, instructions for administration, and/or instructions for mixing the components may also be included in the kit as inserts or labels.
Examples
The present invention will be described in more detail by means of specific examples. The following examples are provided for illustrative purposes and are not intended to limit the invention in any way. Those skilled in the art will readily recognize various non-critical parameters that may be changed or modified to produce substantially the same result. According to at least one assay described herein, the compounds of the examples have been found to be JAK inhibitors.
Example a: in vitro JAK kinase assay
The inhibition activity of JAK1 pathway inhibitors on JAK targets useful in the treatment of cytokine-related diseases or disorders was tested according to the following in vitro assay described in Park et al, analytical Biochemistry 1999,269,94-104. The catalytic domains of human JAK1 (a.a.837-1142), JAK2 (a.a.828-1132) and JAK3 (a.a.781-1124) with an N-terminal His tag were expressed and purified in insect cells using baculoviruses. The catalytic activity of JAK1, JAK2 or JAK3 is determined by measuring the phosphorylation of biotinylated peptides. General purpose medicinePhosphorylated peptides were detected by Homogeneous Time Resolved Fluorescence (HTRF). The IC of the compounds for each kinase was measured in 40. Mu.L of the reaction containing enzyme, ATP and 500nM peptide in 50mM Tris (pH 7.8) buffer containing 100mM NaCl, 5mM DTT and 0.1mg/mL (0.01%) BSA 50 . For 1mM IC 50 The ATP concentration in the reaction was measured to be 1mM. The reaction was allowed to proceed at room temperature for 1 hour and then stopped with 20. Mu.L of 45mM EDTA, 300nM SA-APC, 6nM Eu-Py20 in assay buffer (Perkin Elmer, boston, mass.). Binds to europium-labeled antibody for 40 minutes and the HTRF signal is measured on a Fusion plate reader (Perkin Elmer, boston, MA). The compounds in Table 3 were tested in this assay and showed their IC 50 The values are also found in table 3.
Example B: in vitro JAK kinase assay using melanocyte proliferation assay and 20% cd8+ t cell conditioned medium
The assay investigated in vitro systems using cd8+ T cells and melanocytes. The assay tests whether compound 1 modulates inflammatory responses and how this modulation affects melanocyte biology.
The method comprises the following steps: cd8+ T cell conditioned medium step: normal human cd8+ T cells from peripheral blood were purchased (STEMCELL Technologies). Under activated and non-activated conditions in an Incucyte living cell analysis System (Essen Bioscience) at 37C and 5% CO 2 The cells were then cultured for 48 hours. Activated T cells were cultured in flat bottom 96 well plates (Costar) which were first coated with 10ug/ml anti-CD 3 (BD Biosciences) for 2 hours at room temperature, then washed once with PBS, after which cells were added. Cells were grown in melanocyte growth medium (NHM-GM, matTek) containing 1ug/ml anti-CD 28 (BD Biosciences). Non-activated T cells were treated in the same manner except that they were cultured in separate plates without anti-CD 3 and anti-CD 28. Each T cell plate was treated with compound 1 (1000, 100 or 10 nM) or medium alone at the time of inoculation. Each treatment was performed in triplicate. After 48 hours, all samples (T cells and medium) were transferred to a new V-plate and centrifuged at 1300RPM for 10 minutes. The supernatant was carefully collected from the T cell pellet so as not to interfere with the pellet. Fresh supernatant (CD8+T fines) was used Cell conditioned medium) to treat melanocytes. The remaining amount was stored at-80C and then thawed for protein analysis by Luminex technology (custom Procarta 45-plex, thermo Fisher Scientific). The treated cd8+ T cell pellet was lysed using quantiene sample treatment kit reagents according to kit instructions. Lysates were stored at-80C and then thawed for RNA analysis by Luminex technology using two custom Quantigene 80-Plexes-a total of 150 genes of interest (Thermo Fisher Scientific).
Melanocyte proliferation step: normal human melanocytes (MatTek) isolated from neonatal prepuce epidermis of black donor were purchased. They were thawed and treated at 37C and 5% CO according to the manufacturer's instructions 2 The plates were inoculated down in NHM-GM medium in flat-bottomed 96-well plates (Costar) for 18 hours. When treated with 20% CD8+T cell conditioned medium, they were approximately 10% confluent (25 ul of conditioned medium was added to 100ul of NHM-GM already in the well). Cells were incubated in the Incucyte living cell analysis System (Essen Bioscience) at 37C and 5% CO 2 Incubate for 48 hours, take 5 images every 3 hours per well. After 48 hours, all supernatants were transferred from melanocyte plates to new V-plates and centrifuged at 1300RPM for 10 minutes. The supernatant was collected and stored at-80C and then thawed for protein analysis by Luminex technology (custom Procarta 45-plex, thermo Fisher Scientific). The treated melanocytes were lysed using quantiene sample treatment kit reagents according to the kit instructions. Lysates were stored at-80C and then thawed for RNA analysis of a total of 150 genes of interest (Thermo Fisher Scientific) using two custom Quantigene 80-Plexes by the Luminex technology. The Incucyte images were analyzed using Incucyte software and the percentage of melanocyte confluence per treatment was determined.
Results: FIG. 1 is a graph depicting melanocyte proliferation,% confluence, and time (hours). Compound 1 did not directly alter melanocyte proliferation after transfer of 20% of non-activated T cell medium. The addition of 20% medium from CD3/CD 28-activated human cd8+ T cells to NHM-GM in vitro cultures significantly inhibited melanocyte proliferation compared to non-activated conditions. Co-culture of Compound 1 (1000 nM) with CD3/CD 28-activated human CD8+ T cells partially reversed the inhibited melanocyte proliferation response. This improvement in melanocyte proliferation was statistically significant. Compound 1 added to the inflammatory medium resulted in a significant decrease in spontaneous proliferation compared to the natural melanocyte proliferation rate. Which is a time-dependent decrease in spontaneous natural proliferation reactions.
FIG. 2 is a graph depicting the effect of Compound 1 on a JAK 1-dependent cytokine using T cell compartments, protein T cell medium (pg/ml) and a specific JAK 1-dependent cytokine. Consistent with the previously reported data, co-culturing compound 1 with CD3/CD 28-activated human cd8+ T cells inhibited JAK-STAT-dependent inflammatory cytokines.
FIGS. 3A-3C are graphs depicting the effect of Compound 1 on growth factors and chemokines using T cell compartments, protein T cell medium (pg/ml) and IL-8 (A), HGF (B) and LIF (C). Compound 1, co-cultured with CD3/CD28 activated human cd8+ T cells, reduced several additional mediators that do not have to use JAK1 signaling.
FIG. 4 is a graph depicting the effect of Compound 1 on a JAK 1-dependent cytokine using melanocytes, protein melanocyte medium (pg/ml), and a specific JAK 1-dependent cytokine. Compound 1 reduced JAK-STAT dependent inflammatory cytokines. Some of these cytokines are upregulated by melanocytes under stress conditions (ifnγ, CXCL10, IL-6).
FIGS. 5A-5C are graphs depicting the effect of Compound 1 on growth factors and chemokines obtained using melanocytes, protein melanocyte medium (pg/ml) and IL-8 (A), HGF (B) and LIF (C). IL-8 is upregulated by melanocytes under stress conditions, and this is increased under compound 1 conditions.
Discussion: hepatocyte Growth Factor (HGF)/mesenchymal transition factor (c-MET) signaling protects melanocytes from apoptosis and stimulates their proliferation and movement. After transfer of the activated T cell conditioned medium, the HCF concentration in the melanocyte culture supernatant increases. Compound 1 significantly reduced HGF levels (about 67%) but increased melanocyte proliferation. According to the data (melanocytes and T cells), it is expected that compound 1 reduces HGF expression via signaling mechanisms that would be detrimental to melanocyte proliferation.
Interleukin 8 (IL-8/CXCL 8) has been reported to enhance the proliferative response of melanocytes to a variety of growth factors via binding to its cognate receptors (CXCR 1 and CXCR 2). IL-8 production and CXCR1/2 signaling are both independent of JAK 1. Compound 1 treatment of cd8+ T cells slightly reduced IL-8 concentration (+.22%), but there was a significant increase (+.about.450%) in melanocyte culture. Based on the data (melanocytes and T cells), unexpected upregulation of IL-8 levels in melanocyte cultures was observed, which would enhance the proliferative response.
Leukemia Inhibitory Factor (LIF) increases primary melanocyte proliferation and differentiation. LIF receptor signaling is JAK1 dependent, thus compound 1 is expected to have an adverse effect on melanocyte proliferation. Unexpectedly, melanocyte proliferation was increased. Based on the data (T cells), the mid-target inhibition of LIF does not match the enhancement of melanocyte proliferation response.
Compound 1 down regulates inflammatory cytokines in the cd8+ activated T cell microenvironment. It allows melanocyte proliferation to normalize and also prevents melanocytes from producing inflammatory mediators.
Example C: phase 2 study of Compound 1
Study design
Figure 6 depicts a summary of a phase 2 randomized, double-blind, placebo-controlled dose range study of the efficacy and safety of compound 1. The study was performed using compound 1 for the treatment of participants with non-segmental vitiligo. The study may include a 28 week double blind extension period in vitiligo participants. The participants may include non-segmented vitiligo patients with a depigmented area of 8% or more total BSA, 8T-VASI, 0.5% or more facial BSA, and 0.5F-VASI. Participants will stratified based on total BSA involvement (8-20% and > 20%). In some embodiments, the participants may include non-segmented vitiligo patients with a depigmented area of 5% or more total BSA, including 5T-VASI, 0.5% or more facial BSA, and 0.5F-VASI. Participants will stratified based on total BSA involvement (8-20% and > 20%). The study may include men and women aged 18 to 75 years.
A 24 week placebo-controlled double-blind period would have approximately 160 participants randomly allocated to 1 of the 3 treatment groups (dose a, dose B or dose C) or placebo group at 1:1:1:1. A 28 week double blind extension would include all participants who successfully completed the placebo-controlled period. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about dose B based on the free base. In some embodiments, the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about dose C based on the free base. The primary endpoint was the percentage change in total vitiligo area score index (T-VASI) from baseline at week 24, and the key secondary endpoint was the proportion of participants who achieved T-VASI50 at week 24. In some embodiments, the results of this study will help inform the choice of dose to be evaluated in the phase 3 study.
In some embodiments, the criteria that exclude participants from the study include any of the following: other skin depigmentation disorders (e.g., albinism, pityriasis albus, leprosy, post-inflammatory hypopigmentation, progressive plaque hypopigmentation (progressive macule hypomelanosis), anemic mother-spots, chemical white skin, and tinea versicolor); thyroid function was uncontrolled upon screening as determined by the investigator (note If the participant has a history of thyroid disease and is receiving treatment, the participant must employ a stable thyroid regimen at least three months prior to day 1); treatment with laser or light-based (phototherapy), including the use of a sun bed within 8 weeks prior to day 1; dihydroxyacetone (typically present in a sunless product) was used within 4 weeks prior to day 1; the monobenzyl ethers of hydroquinone, which are currently or in the past used as depigmenting agents, comprise(Mo Nuoben zone); melanocyte-keratinocyte transplantation surgery (MKTP) or other history of vitiligo surgery; spontaneous and significant pigment regeneration (e.g., pigment regeneration without any treatment, and significant number determined by the investigator) within 6 months prior to screening; pregnancy or under examinationWomen who are concerned about pregnancy or lactation; treatment of a medical history of vitiligo or any other inflammatory disorder failure with any systemic or local JAK or TYK2 inhibitor (e.g., pontine, tofacitinib, baratinib, non-golitinib, letatinib, pacatinib, abatinib, bricitinib; anticoagulants or drugs known to cause thrombocytopenia are currently used; hepatitis: evidence of HBV or HCV infection or reactivation risk (participants cannot be positive for HBV DNA, HCV RNA, HBsAg or anti-hepatitis b core antibodies; but participants who had no history of HBV infection previously and had been vaccinated with HBV vaccine and had positive antibodies against HBV surface antigen (anti-hbs+) as the sole evidence of previous exposure can participate in the study); known HIV infection; hypersensitivity or severe reactions to compound 1 or the excipients of compound 1 are known; researchers believe that participants are unlikely or impossible to follow the dosing schedule and study evaluations; and at the discretion of the investigator, any condition that would interfere with complete participation in the study (including administration of study medication and participation in the study visit required); constitutes a significant risk to the participants; or interfere with interpretation of study data.
In some embodiments, the criteria that exclude participants from the study include participants having a history of a concurrent disorder or other disease, including any of the following: a history of thrombocytopenia, coagulation disorders, platelet dysfunction or thrombotic events; any clinically significant medical condition other than vitiligo, as determined by a researcher, which is not adequately controlled by appropriate treatment or may interfere with the course, severity or assessment of this study; any other active skin disease or condition that may interfere with the course, severity, or assessment of this study; based on the clinical assessment of the investigator, any bacterial, fungal or viral infection makes the participants candidates unsuitable for research; current history of herpes zoster infection, disseminated herpes simplex or history of herpes zoster; a history of malignancy including melanoma, lymphoma, and leukemia within 5 years prior to day 1, except for successfully treated non-metastatic skin squamous cell carcinoma, basal cell carcinoma, or localized cervical carcinoma in situ; and albinism.
In some embodiments, the criteria that exclude the participants from the study include receiving the drug or study drug within the following interval prior to day 1 (first administration of study drug): for any local or systemic JAK or TYK2 inhibitor, <12 weeks or 5 half-lives (if known), whichever is longer; for any investigational or experimental treatment, <12 weeks or 5 half-lives (if known), whichever is longer; for systemic immunosuppression or immunomodulation biopharmaceuticals (e.g., adalimumab (egadalimab), etanercept, infliximab, golimumab (golimumab), cetuximab (certolizumab), ulimumab (ustekumab), secukinumab (secukinumab), budaimumab (brodalumab), iximab Bei Shan (ixekizumab), risenzumab (risenkizumab), gulek You Shan antibody (guselkuumab), bimetaizumab (bimekizumab), icalizumab (iscaliumab), bei Maiji mab (bermekimab), rituximab (rituximab), anakinra), 12 weeks or 5 half-lives if known; for live vaccination, or for a period of 6 weeks scheduled to vaccinate during the course of the study or after the last dose of study medication, <6 weeks; for systemic immunosuppression or immunomodulation small molecule drugs (e.g., oral or injected corticosteroids, methotrexate, cyclosporine, dapsone, azathioprine) <4 weeks; for any oral or topical PDE-4 inhibitor (e.g., aplastt, clenbuterol) <3 weeks; for any topical drug for vitiligo lesions, <2 weeks; for any OTC therapy to treat vitiligo, <2 weeks; for strong and moderate systemic CYP3A4 inhibitors and strong systemic CYP3A4 inducers within 2 weeks or 5 half-lives (if known), the longer of them is preferred. Examples include, but are not limited to, the following: erythromycin, rifampicin/Lei Faping (rifampicin), ciprofloxacin, some azole antifungals (e.g., ketoconazole, fluconazole), nefazodone, san john's word, diltiazem, mibezil, verapamil, grapefruit/grapefruit juice, and lime; and for antiplatelet therapy, <1 week.
In some embodiments, the criteria that exclude participants from the study include the use of systemic immunosuppressive or immunomodulatory small molecule drugs, note: allowing the use of corticosteroid inhalants and intranasal sprays; if the investigator and sponsor deems acceptable, oral corticosteroids are allowed to treat non-dermatological disorders (e.g., asthma exacerbation, bronchitis) for no more than 7 days; and allowing the use of topical corticosteroids for the treatment of skin disorders other than vitiligo (e.g. atopic dermatitis or psoriasis) in areas not receiving vitiligo treatment (the total BSA involvement of other skin disorders must not exceed 10% except in the vitiligo treated areas).
In some embodiments, the criteria that exclude participants from the study include the use of systemic immunosuppressive or immunomodulatory small molecule drugs, note: at the discretion of the investigator, low doses of acetylsalicylic acid (.ltoreq.100 mg QD) were allowed for cardiovascular prophylaxis purposes.
In some embodiments, the criteria that exclude participants from the study include evidence of active or latent or undertreated mycobacterium tuberculosis (i.e., TB) infection defined by at least one of: positive results were performed 12 weeks before day 1 or within 12 weeks The TB Gold In-Tube test (QFT-GIT) or positive Mantoux/PPD tuberculin skin test is exclusionary and requires a negative test to qualify unless other criteria described herein apply (note: participants who suggest a history of BCG vaccination should be tested using QFT-GIT because Mantoux/PPD tuberculin skin test may be positive for vaccination (note: if the participant has previously received a recorded adequate course of treatment for latent or active TB infection, no QFT-GIT or Mantoux/PPD tuberculin skin test is required)); history of untreated or inappropriately treated latent or active TB infection; if the participant has previously been resistant to latency (in primary multi-drug TBRate of<5% of sites received adequate treatment using isonicotinic tincture for 9 months or acceptable alternatives) or active (acceptable multi-drug regimen) TB infection, no QFT-GIT or Mantoux/PPD tuberculin skin test was required, but chest radiographs or other appropriate diagnostic images were required within 3 months of day 1 (note: in order to be considered as meeting the study conditions, radiographs must be determined by qualified radiologists to be negative for active tuberculosis infection, and documented and negative chest radiography results for adequate treatment of TB must be obtained prior to day 1; and participants currently undergoing treatment for active TB infection.
In some embodiments, the criteria for the participants excluded from the study include participants with laboratory values at screening defined in table 4:
TABLE 4 Table 4
Example D: in vitro JAK kinase assay using melanocyte proliferation assay and 20% cd8+ t cell conditioned medium
The assay investigated in vitro systems using cd8+ T cells and melanocytes. The assay tested whether and how the modulation of the inflammatory response by pontine affects melanocyte biology.
The method comprises the following steps: cd8+ T cell conditioned medium step: normal human cd8+ T cells from peripheral blood were purchased (STEMCELL Technologies). Under activated and non-activated conditions in an Incucyte living cell analysis System (Essen Bioscience) at 37C and 5% CO 2 The cells were then cultured for 48 hours. Activated T cells were cultured in flat bottom 96 well plates (Costar) which were first coated with 10ug/ml anti-CD 3 (BD Biosciences) for 2 hours at room temperature, then washed once with PBS, after which cells were added. Cells were grown in melanocyte growth medium (NHM-GM, matTek) containing 1ug/ml anti-CD 28 (BD Biosciences). Non-activated T cells were treated in the same manner except that they were cultured in separate plates without anti-CD 3 and anti-CD 28. Each T cell plate was seeded with When treated with ponatinib (1000, 100 or 10 nM) or medium alone. Each treatment was performed in triplicate. After 48 hours, all samples (T cells and medium) were transferred to a new V-plate and centrifuged at 1300RPM for 10 minutes. The supernatant was carefully collected from the T cell pellet so as not to interfere with the pellet. Melanocytes were treated with fresh supernatant (cd8+ T cell conditioned medium). The remaining amount was stored at-80C and then thawed for protein analysis by Luminex technology (custom Procarta 45-plex, thermo Fisher Scientific). The treated cd8+ T cell pellet was lysed using quantiene sample treatment kit reagents according to kit instructions. Lysates were stored at-80C and then thawed for RNA analysis by Luminex technology using two custom Quantigene 80-Plexes-a total of 150 genes of interest (Thermo Fisher Scientific).
Melanocyte proliferation step: normal human melanocytes (MatTek) isolated from neonatal prepuce epidermis of black donor were purchased. They were thawed and treated at 37C and 5% CO according to the manufacturer's instructions 2 The plates were inoculated down in NHM-GM medium in flat-bottomed 96-well plates (Costar) for 18 hours. When treated with 20% CD8+T cell conditioned medium, they were approximately 10% confluent (25 ul of conditioned medium was added to 100ul of NHM-GM already in the well). Cells were incubated in the Incucyte living cell analysis System (Essen Bioscience) at 37C and 5% CO 2 Incubate for 48 hours, take 5 images every 3 hours per well. After 48 hours, all supernatants were transferred from melanocyte plates to new V-plates and centrifuged at 1300RPM for 10 minutes. The supernatant was collected and stored at-80C and then thawed for protein analysis by Luminex technology (custom Procarta 45-plex, thermo Fisher Scientific). The treated melanocytes were lysed using quantiene sample treatment kit reagents according to the kit instructions. Lysates were stored at-80C and then thawed for RNA analysis of a total of 150 genes of interest (Thermo Fisher Scientific) using two custom Quantigene 80-Plexes by the Luminex technology. The Incucyte images were analyzed using Incucyte software and the percentage of melanocyte confluence per treatment was determined.
Results: fig. 7 is a graph depicting melanocyte proliferation,% confluence, and time (hours). The addition of 20% medium from CD3/CD 28-activated human cd8+ T cells to NHM-GM in vitro cultures significantly inhibited melanocyte proliferation compared to non-activated conditions. Co-culture of ponatinib (1000 nM) with CD3/CD28 activated human CD8+ T cells appears to completely reverse the inhibited melanocyte proliferation response. This improvement in melanocyte proliferation was statistically significant. The addition of ponatinib to the inflammatory medium resulted in a significant decrease in spontaneous proliferation compared to the natural melanocyte proliferation rate. Which is a time-dependent decrease in spontaneous natural proliferation reactions.
FIG. 8 is a graph depicting the effect of ponatinib on a JAK 1-dependent cytokine using a T cell compartment, protein T cell medium (pg/ml) and a specific JAK 1-dependent cytokine. Consistent with the previously reported data, the co-culture of pontinib with CD3/CD28 activated human CD8+ T cells inhibited the JAK-STAT dependent inflammatory cytokines, but the inflammatory cytokines IL-2 were upregulated (seemed to have no effect on inflammatory cytokines IL-4, IL-5 (note that IL-5 is JAK 2/2) and IL-6).
FIGS. 9A-9B are graphs depicting the effect of ponatinib on growth factors and chemokines obtained using T cell compartments, protein T cell medium (pg/ml) and IL-8 (A) and LIF (B). Ponatinib co-cultured with CD3/CD28 activated human cd8+ T cells reduced several additional mediators that do not have to use JAK1 signaling.
FIG. 10 is a graph depicting the effect of ponatinib on a JAK 1-dependent cytokine using melanocytes, protein melanocyte medium (pg/ml), and a specific JAK 1-dependent cytokine. Ponatinib reduced JAK-STAT dependent inflammatory cytokines (seemed to have no effect on inflammatory cytokines IL-13 and IL-10). Some of these cytokines are upregulated by melanocytes under stress conditions (ifnγ, CXCL10, IL-6).
FIGS. 11A-11B are graphs depicting the effect of ponatinib on growth factors and chemokines obtained using melanocytes, protein melanocyte medium (pg/ml) and IL-8 (A) and LIF (B). IL-8 is upregulated by melanocytes under stress conditions and decreased under pontine conditions.
Discussion: interleukin 8 (IL-8/CXCL 8) has been reported to enhance the proliferative response of melanocytes to a variety of growth factors by binding to its cognate receptors (CXCR 1 and CXCR 2). IL-8 production and CXCR1/2 signaling are both independent of JAK 1. Treatment of cd8+ T cells with pontinib slightly reduced IL-8 concentration.
Leukemia Inhibitory Factor (LIF) increases primary melanocyte proliferation and differentiation. LIF receptor signaling is JAK1 dependent, thus, rucotinib is expected to have an adverse effect on melanocyte proliferation. Unexpectedly, melanocyte proliferation was increased. Based on the data (T cells), the mid-target inhibition of LIF does not match the enhancement of melanocyte proliferation response.
The poncirtinib down regulates inflammatory cytokines in the cd8+ activated T cell microenvironment. It allows melanocyte proliferation to normalize and also prevents melanocytes from producing inflammatory mediators.
Various modifications of the application in addition to those described herein will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference cited in this application, including all patents, patent applications, and publications, is incorporated by reference in its entirety.

Claims (11)

1. A method for treating vitiligo in a subject, the method comprising administering to the subject a therapeutically effective amount of a JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof.
2. The method of claim 1, wherein the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is selective for JAK1 over JAK2, JAK3 and Tyk2.
3. The method of claim 1, wherein the JAK1 pathway inhibitor is 4- [3- (cyanomethyl) -3- (3 ',5' -dimethyl-1 h,1'h-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2, 2-trifluoro-1-methylethyl ] benzamide or a pharmaceutically acceptable salt thereof.
4. The method of claim 1, wherein the JAK1 pathway inhibitor is 4- [3- (cyanomethyl) -3- (3 ',5' -dimethyl-1 h,1'h-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2, 2-trifluoro-1-methylethyl ] benzamide phosphate.
5. The method of any one of claims 1-4, wherein the vitiligo is non-segmental vitiligo.
6. The method of any one of claims 1-5, wherein the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about 5mg to about 95mg based on free base.
7. The method of any one of claims 1-5, wherein the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in a daily dose of about 15mg, about 45mg, 75mg, or about 90mg on a free base basis.
8. The method of any one of claims 1-7, wherein the JAK1 pathway inhibitor or pharmaceutically acceptable salt thereof is administered in combination with an additional therapeutic agent.
9. The method of claim 8, wherein the additional therapeutic agent comprises a Janus kinase inhibitor.
10. The method of claim 9, wherein the Janus kinase inhibitor comprises pontinib or a pharmaceutically acceptable salt thereof.
11. The method of any one of claims 1-10, wherein the administering comprises administering the JAK1 pathway inhibitor or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable carrier or excipient.
CN202180082167.5A 2020-12-08 2021-12-08 JAK1 pathway inhibitors for the treatment of vitiligo Pending CN116710088A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/122,574 2020-12-08
US202163246688P 2021-09-21 2021-09-21
US63/246,688 2021-09-21
PCT/US2021/062419 WO2022125670A1 (en) 2020-12-08 2021-12-08 Jak1 pathway inhibitors for the treatment of vitiligo

Publications (1)

Publication Number Publication Date
CN116710088A true CN116710088A (en) 2023-09-05

Family

ID=87839737

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180082167.5A Pending CN116710088A (en) 2020-12-08 2021-12-08 JAK1 pathway inhibitors for the treatment of vitiligo

Country Status (1)

Country Link
CN (1) CN116710088A (en)

Similar Documents

Publication Publication Date Title
US20190060311A1 (en) Compositions and methods for treatment of vitiligo
US11833152B2 (en) JAK1 pathway inhibitors for the treatment of cytokine-related disorders
CN112423759A (en) Treatment of hidradenitis suppurativa with JAK inhibitors
US9849132B2 (en) Products and pharmaceutical compositions
CN117693339A (en) JAK1 pathway inhibitors for the treatment of prurigo nodularis
KR101802011B1 (en) 2,4 substituted pyrimidinediamines for use in discoid lupus
US11957661B2 (en) JAK1 pathway inhibitors for the treatment of vitiligo
JP6563998B2 (en) Treatment plan
JP2023506118A (en) Use of JAK1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (LP)
CN116710088A (en) JAK1 pathway inhibitors for the treatment of vitiligo
US20240058343A1 (en) Treatment of urticaria using jak inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination