Methods of treating sjogren's syndrome using bruton's tyrosine kinase inhibitors

文档序号:411080 发布日期:2021-12-17 浏览:17次 中文

阅读说明:本技术 使用布鲁顿酪氨酸激酶抑制剂治疗舍格伦综合征的方法 (Methods of treating sjogren's syndrome using bruton's tyrosine kinase inhibitors ) 是由 S·巴塔查里亚 B·比耶特 M·卡班斯基 B·岑尼 S·德巴克 M·考尔 A·基希卡尔 于 2020-05-20 设计创作,主要内容包括:本披露涉及使用具有式(I)的化合物或其药学上可接受的盐治疗舍格伦综合征的方法。本文还披露了用于治疗舍格伦综合征患者的具有式(I)的化合物或其药学上可接受的盐,以及用于所披露的用途和方法的药物、给药方案、药物配制品、剂型和试剂盒。(The present disclosure relates to methods of treating sjogren's syndrome using compounds having formula (I) or a pharmaceutically acceptable salt thereof. Also disclosed herein are compounds having formula (I) or a pharmaceutically acceptable salt thereof, for use in treating patients with sjogren's syndrome, as well as medicaments, dosing regimens, pharmaceutical formulations, dosage forms and kits for the disclosed uses and methods.)

1. A compound having the formula (I):

or a pharmaceutically acceptable salt thereof, for use in treating sjogren's syndrome (SjS) in a subject in need of such treatment, wherein the daily dose of the compound having formula (I) is from about 10mg to about 200 mg.

2. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the daily dose is from about 10mg to about 100 mg.

3. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the daily dose is about 100 mg.

4. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the daily dose is about 50 mg.

5. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the daily dose is about 35 mg.

6. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the daily dose is about 25 mg.

7. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the daily dose is about 20 mg.

8. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the compound of formula (I) is administered once daily at a dose of about 10mg, about 35mg, about 50mg, or about 100 mg.

9. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to claim 1, wherein the compound of formula (I), or a pharmaceutically acceptable salt thereof, is administered twice daily at a dose of about 10mg, about 25mg, about 50mg, or about 100 mg.

10. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to any one of the preceding claims, wherein the subject has moderate to severe SjS.

11. The compound having formula (I), or a pharmaceutically acceptable salt thereof, for use according to any one of claims 1-10, wherein the subject is selected according to at least one of the following criteria:

a) (ii) the ESSPRI score of the subject is ≧ 5 prior to treatment with the compound having formula (I);

b) the subject's 8 defined domains from biological, hematological, joint, skin, gland, lymph node pathology, kidney and constitutional features have a weighted score-based ESSDAI ≧ 5 prior to treatment with the compound having formula (I).

12. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to any one of the preceding claims, wherein the subject is an adult human.

13. The compound of formula (I) or a pharmaceutically acceptable salt thereof for use according to any one of the preceding claims, wherein the subject achieves at least one of the following at week 12 or 24 of treatment:

a) a decrease in the ESSPRI score; and/or

b) A decrease in the ESSDAI score.

14. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to any one of the preceding claims, wherein the subject achieves a sustained response as measured by espri or EDSSDAI at week 5 after completion of the treatment.

15. The compound of formula (I), or a pharmaceutically acceptable salt thereof, for use according to any preceding claim, wherein the compound of formula (I), or a pharmaceutically acceptable salt thereof, is configured in a pharmaceutical formulation, and wherein the pharmaceutical formulation further comprises a pharmaceutically acceptable carrier.

16. The compound having the formula (I), or a pharmaceutically acceptable salt thereof, for use according to any one of claims 1-15, wherein T of the compound having the formula (I), or a pharmaceutically acceptable salt thereofmaxFrom about 0.5 to about 3 hours.

Technical Field

The present disclosure relates to the treatment of sjogren's syndrome using Bruton's Tyrosine Kinase (BTK) inhibitors: (syndrome).

Background

Sjogren's syndrome (SjS) is a systemic autoimmune disease of unknown etiology characterized by lymphoid infiltration and progressive destruction of exocrine glands (Brito-Zer Lolo P. et al, (2016) Treating the undermining Pathopathy of PrimarySyndrome, Recent Advances and Future Prospecs [ treatment of the underlying pathophysiology of Primary Sjogren Syndrome: recent progress and future prospects]Drugs]Page 1601 + 1623).

Although the disease primarily affects the lacrimal and salivary glands, the inflammatory process can target any organ, with about 15% of patients exhibiting severe extra-glandular manifestations (Baldini c. et al (2014) Primarysyndrome as a multi-organ disease:impact of the serological profile on the clinical presentation of the disease in a large heart of Italian Patents [ Primary Sjogren syndrome as a multiple organ disease: effect of serological characteristics on the clinical manifestations of the disease in a large cohort of Italian patients]Rheumatology (Oxford) [ rheumatology (Oxford)]Pages 839-44). The most common major feature of clinical manifestations is the exocrine lesions of the saliva and lacrimal glands, manifested as dry mouth and eyes. However, symptoms can vary widely, ranging from dryness, and also including musculoskeletal pain and fatigue affecting almost all patients, to severe, extraglandular, and systemic involvement (characterized by periepithelial lymphocyte infiltration and immune complex deposition). SjS is that destruction of the exocrine gland epithelium results in autoreactive B and T cells (Brito-Zer Lolo n P. et al, (2016) Treating the undermining Pathopathy of PrimarySyndrome, Recent Advances and Future Prospecs [ treatment of the underlying pathophysiology of Primary Sjogren Syndrome: recent progress and future prospects]Drugs]Page 1601 + 1623). The high prevalence of autoantibodies, particularly antibodies to Ro/SSA, even at an early stage suggests that autoreactive B cells are involved in the pathogenesis of SjS (noctrurne g, et al, (2018) B cells in the pathogenesis of primaryB cells in syndrome [ primary Sjogren's syndrome pathogenesis]Nat Rev Rheumatology review]Page 133-145).

The B cell pathology also results in an increased risk of malignant transformation, with a 10-fold increase in the lifetime risk of B cell lymphoma in 5% of SjS patients (Baldini c. et al, (2014) Primarysynthetic as a multi-organ disease, an impact of the clinical profile on the clinical presentation of the disease in a large family of Italian patents [ primary sjogren's syndrome as a multi-organ disease: large queueEffect of serological characteristics on the clinical manifestations of the disease in Italian patients]Rheumatology](Oxford) pages 839-44). It is estimated that the prevalence of SjS ranges from 0.3 to 1 person per 1000 persons (Qin B. et al (2015) Epidemiology of primary)Epidemiology of syndrome: systematic review and meta-analysis of Primary Sjogren syndrome]Ann. Rheum. Dis. [ year of rheumatic diseases]Pages 1983-9) and is second only to rheumatoid arthritis. The disease affects primarily women, with a female/male ratio of 9:1, and can occur at any age. SjS the main effect of symptoms is a severe impact on quality of life and productivity, usually caused by disabling fatigue associated with the disease (Mariette x. et al (2018) Primary)Syndrome [ primary sjogren's Syndrome]N.engl.j.med. [ new england journal of medicine]Pages 931 and 939). There are also many potential severe systemic complications, including arthritis, cutaneous vasculitis, peripheral neuropathy, glomerulonephritis, interstitial nephritis, cholangiogenic cholangitis, obstructive bronchiolitis, and others, involving multiple organ systems, and affecting 20% -40% of patients (Seror r et al (2014) outcom measles for primary maryA comprehensive review [ outcome measure of primary sjogren's syndrome: general overview]J.autoimmun. [ J.Immunity journal]Pages 51-6).

The clinical characteristics of sjogren's syndrome can be divided into medically valuable manifestations and patient symptomatic manifestations. Currently, there is no single assessment tool that can capture SjS these two types of clinically manifested disease activity. Thus, the "european antirheumatic alliance (EULAR) Sjogren Syndrome (SS) patient reporting index (ESSPRI)" and EULAR SS disease activity index (ESSDAI) are widely accepted and validated to measure SjS symptoms and systemic manifestations (franciscini f. et al, (2017), BMC Medicine [ BMC medical ],15: 69).

With respect to the current therapeutic prospects, there is no internationally recognized SjS systemic therapy. In the case of dry mouth and eyes, treatment of SjS patients is limited to symptomatic care. Steroids and typical DMARDs are largely ineffective and no drug intervention is available to combat severe disabling fatigue. The lack of effective treatment options highlights the need for new therapeutic approaches to evaluate this highly debilitating disease. Since the pattern of B cell autoreactions is somewhat similar to that of systemic lupus erythematosus and rheumatoid arthritis, SjS glandular and extraglandular manifestations of B cell depletion therapy with the anti-CD 20 monoclonal antibody (mAb) rituximab and treatment of lymphomas with varying degrees of success have recently been evaluated. However, this method is not currently approved as SjS treatment. Insufficient efficacy of rituximab may be associated with incomplete depletion of B cells in affected tissues (Brito-Zer Lo n P et al (2016) Treating the undermining Pathopathy of PrimarySyndrome, Recent Advances and Future Prospecs [ treatment of the underlying pathophysiology of Primary Sjogren Syndrome: recent progress and future prospects]Drugs]Page 1601 + 1623).

Despite the existing treatments for SjS, there remains a great medical need to provide SjS subjects with new treatment options.

Disclosure of Invention

It is an object of the present invention to provide a novel method of treating sjogren's syndrome disease in a subject, said method comprising administering to said subject in need of such treatment a therapeutically effective amount of NI- (3- (6-amino-5- (2- (NI-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide, or a pharmaceutically acceptable salt thereof.

Accordingly, disclosed herein is a method of treating sjogren's syndrome (SjS), comprising administering to a subject in need of such treatment a daily dose of from about 0.5mg to about 600mg, preferably a daily dose of from about 10mg to about 200mg, or more preferably a daily dose of from about 10mg to about 100mg of NI- (3- (6-amino-5- (2- (NI-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide, or a pharmaceutically acceptable salt thereof.

Also disclosed are NI- (3- (6-amino-5- (2- (NI-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide; or a pharmaceutically acceptable salt thereof, for use in therapy SjS, wherein NI- (3- (6-amino-5- (2- (NI-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is administered in a daily dose from about 0.5mg to about 600mg, preferably in a daily dose from about 10mg to about 200mg, and most preferably in a daily dose from about 10mg to about 100 mg.

Drawings

FIG. 1 shows a schematic view of a: blood concentration-time course of Compound (I) after Single ascending dose of 0.5mg to 600mg

FIG. 2Multiple ascending doses 10mg to 400mg q.d. blood concentration-time course of Compound (I) after administration

FIG. 3Blood concentration-time course of Compound (I) after multiple escalating doses of 100mg b.i.d. to 200mg b.i.d. -%

FIG. 4: food effect observed after a single oral dose of 60mg of a compound having formula (I)

FIG. 5: arithmetic mean (SD) percentage of BTK occupancy in peripheral blood after a single dose of a compound of formula (I)

FIG. 6: percent inhibition of basophil activation on day 12 of multiple ascending doses of the compound of formula (I) relative to the total daily dose of the compound of formula (I)

FIG. 7: wheal size reduction in multiple incremental dose skin prick tests

Detailed Description

Bruton's Tyrosine Kinase (BTK) is a cytoplasmic tyrosine kinase and is a member of the TEC kinase family. BTK is found in cells of the adaptive and innate immune systems,including B cells, macrophages, basophils, mast cells and platelets. BTK is essential for signaling through both fcepsilon receptors (fcepsilon R1 for IgE) and activated fcgamma receptors (fcgamma R for IgG) as well as B cell antigen receptors (BCR). BTK inhibition has been shown to be an effective concept for the treatment of B cell malignancies. Covalent BTK inhibitor ibrutinibAcatinibAnd ZanbutiniApproved for the treatment of certain B-cell malignancies (Thompson PA et al, (2018) Bruton's tyrosine kinase inhibitors: first and second generation agents for tissues with Chronic Lymphocytic Leukemia (CLL) [ Bruton tyrosine kinase inhibitors: first and second generation agents for patients with Chronic Lymphocytic Leukemia]Expert Opin Investig Drugs [ Experimental drug experts opinion ]]Pages 31-42). BTK inhibition has shown promising efficacy for B cell autoimmunity in preclinical and clinical studies (Tan SL. et al, (2013) Targeting the SYK-BTK antigens for the treatment of immunological and hematological disorders [ Targeting the SYK-BTK axis for the treatment of immunological and hematological diseases: recent progress and therapeutic prospects]Thers [ pharmacology and therapeutics ]]Pages 294-; whang J.A. et al (2014) Bruton's tyrosine kinase inhibitors for the treatment of rheumatoid arthritis]Drug discovery]Pages 1200-4; satterhwaite A.B. (2017) Bruton's Tyrosine Kinase, a Component of B Cell Signaling Pathways, Has Multiple loops in the pathway of Lupus [ B Cell Signaling pathway Component Bruton Tyrosine Kinase Has Multiple functions in Pathogenesis of Lupus erythematosus]Front Immunol [ immunological frontier]Page 1986; rip J. et al(2018) The Role of Bruton's Tyrosine Kinase in Immune Cell Signaling and Systemic Autoimmunity]Crit.rev.immunol. [ immunological review ]]Pages 17-62). Therefore, inhibition of BTK is an attractive therapeutic concept for the treatment of various autoimmune and chronic inflammatory diseases, including rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, chronic urticaria, atopic dermatitis, asthma, and primary sjogren's syndrome (Tan SL, Liao C, Lucas MC et al (2013) Targeting the SYK-BTK axis for the treatment of immunological and hematological disorders: recent progress and treatment prospects]Thers [ pharmacology and therapeutics ]]Pages 294-; a Brang JA, Chang BY (2014) Bruton's tyrosine kinases inhibitors for the treatment of rheumatoid arthritis [ Bruton's tyrosine kinase inhibitors]Drug discovery]Pages 1200-4).

Furthermore, elevated levels of BTK in circulating B cells were shown in a significant proportion of SjS Patients, associated With higher serum Rheumatoid Factor (RF) levels (Corneth OBJ et al (2017) Enhanced Bruton's Tyrosine Activity in Peripheral Blood BLymphocytes From Patients With Autoimmune Disease Enhanced Bruton's Tyrosine Kinase Activity P. page 1313-1324).

N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide, or a pharmaceutically acceptable salt thereof, is a BTK inhibitor, referred to herein as a compound having the formula (I):

or a pharmaceutically acceptable salt thereof.

The compounds are described in WO 2015/079417 (attorney docket No. PAT056021-WO-PCT) filed on 4.6.2015. The compounds are selective, potent, irreversible covalent inhibitors of Bruton's Tyrosine Kinase (BTK) and are useful in BTK-mediated diseases or disorders.

Thus, we have now devised a dosing regimen for treating a SjS patient with the compound N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide, or a pharmaceutically acceptable salt thereof.

Defining:

for the purpose of explaining the present specification, the following definitions will apply and, where appropriate, terms used in the singular will also include the plural and vice versa.

The phrase "pharmaceutically acceptable" as used herein refers 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.

Any formula given herein is also intended to represent the unlabeled form as well as the isotopically labeled form of the compound. Isotopically labeled compounds have the structure represented by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Isotopes that can be incorporated into compounds of the present disclosure include isotopes of, for example, hydrogen, carbon, nitrogen, oxygen, fluorine, iodine, and chlorine, such as3H、11C、13C、14C、15N、18F and36and (4) Cl. Thus, it is to be understood that the present disclosure includes incorporation of any of the foregoing isotopes (including, for example, radioisotopes (such as3H and14C) or in the presence of a non-radioactive isotope (such as2H and13C) those of (a). Such isotopically labeled compounds are useful in metabolic studies (using14C) Reaction kinetics study (e.g. with2H or3H) Detection or imaging techniques (e.g. Positron Emission Tomography (PET) or single photon emission meters)Computed Tomography (SPECT), including drug or substrate tissue distribution assays), or for radiation treatment of a patient. In particular, it is possible to use, for example,18f or labeled compounds may be particularly desirable for PET or SPECT studies. Isotopically labeled compounds can generally be prepared by conventional techniques known to those skilled in the art, for example by using an appropriate isotopically labeled reagent in place of an unlabeled previously used reagent.

The term "pharmaceutical composition" as used herein refers to a product obtained by using or mixing or combining more than one active ingredient. It is to be understood that the pharmaceutical compositions used herein include fixed and non-fixed combinations of active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound of formula (I) or a pharmaceutically acceptable salt thereof and one or more combination partners, are administered to a patient simultaneously as a single entity or dosage form. In this case, the term refers to a fixed dose combination in one unit dosage form (e.g., capsule, tablet or sachet). The terms "non-fixed combination" or "kit of parts" both denote that the active ingredients, e.g. a compound of the present disclosure and one or more combination partners and/or one or more auxiliaries, are administered sequentially as separate entities simultaneously, simultaneously or without specific time constraints or separately co-administered to a patient, wherein such administration provides therapeutically effective levels of both compounds in the patient's body, especially in case of effective doses at such time intervals as to allow the combination partners to show a synergistic effect, e.g. additive or synergistic effect. The term "non-fixed combination" also applies to a mixture therapy, e.g. the administration of three or more active ingredients. Thus, the term "non-fixed combination" especially defines an application, use, composition or formulation in the sense that the compounds described herein can be administered independently of each other (i.e. simultaneously or at different time points). It is to be understood that the term "non-fixed combination" also encompasses the use of a single agent together with one or more fixed combination products, wherein each separate formulation has a different amount of the active ingredient. It is further understood that the combination products described herein and the term "non-fixed combination" encompass active ingredients (including the compounds described herein) wherein the combination partners are administered in pharmaceutical dosage forms or pharmaceutical formulations which are sold completely separately, also independently of one another. Instructions for use of the non-fixed combination are or may be provided in a package (e.g., leaflet, etc.) or other information provided to the physician and/or medical personnel. The individual formulations or the portions of the formulation, product or composition may then be applied simultaneously or chronologically staggered, i.e., the individual portions of the kit of parts may be applied at different time points and/or any portion of the kit of parts may be applied at the same or different time intervals. In particular, the time intervals of administration are chosen such that the effect on the treated disease in the combined use of the parts is larger/superior to the effect which would be obtained by the use of the compound of formula (I) alone; thus, the compounds used in the pharmaceutical combinations described herein have a combined activity. The ratio of the total amount of the compound of formula (I) to the second agent to be administered as a pharmaceutical combination may be varied or adjusted in order to better meet the needs of the particular patient sub-population to be treated or the needs of the individual patient, which needs may be due to, for example, the age, sex, body weight, etc. of the patient.

As used herein, the terms "co-administration" or "combined administration" and the like are intended to encompass administration of one or more compounds described herein with a selected combination partner to a single subject (e.g., a patient or subject) in need thereof, and are intended to include treatment regimens in which the compounds are not necessarily administered by the same route of administration and/or concurrently.

The term "pharmaceutical composition" is defined herein to mean a mixture (e.g., a solution or emulsion) containing at least one active ingredient or therapeutic agent to be administered to a warm-blooded animal (e.g., a mammal or a human) for the prevention or treatment of a particular disease or condition affecting said warm-blooded animal.

The term "therapeutically effective amount" of a compound of the present disclosure (i.e., a compound having formula (I), or a pharmaceutically acceptable salt thereof) refers to an amount of a compound of the present disclosure that will elicit a biological or medical response in a subject (subject patient), e.g., a reduction or inhibition of the activity of an enzyme or protein in the subject (subject patient), or ameliorate a symptom, alleviate a condition, slow or delay the progression of a disease or prevent a disease, etc. The therapeutically effective dose of the compound, pharmaceutical composition or combination thereof depends on the species, body weight, age, sex and individual condition of the patient, the disorder or disease to be treated or its severity. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each active ingredient which is necessary to prevent, treat or inhibit the progression of the disorder or disease.

The frequency of dosage may vary depending on the compound used and the particular condition being treated or prevented. In general, it is preferred to use a minimum dose sufficient to provide an effective therapy. The effectiveness of a treatment in a patient can generally be monitored using assays appropriate for the condition being treated or prevented, which assays will be familiar to those of ordinary skill in the art.

As used herein, the term "carrier" or "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, binders, excipients, disintegrants, lubricants, sweeteners, flavoring agents, dyes, and the like, and combinations thereof, as would be understood by one of skill in the art (see, e.g., Remington's pharmaceutical Sciences, 18 th edition, Mack Printing Company, 1990, 1289-1329). Unless any conventional carrier is incompatible with the active ingredient, it is contemplated that it may be used in therapeutic or pharmaceutical compositions.

As used herein, the term "subject" refers to an animal. Typically, the animal is a mammal. Subjects also refer to, for example, primates (e.g., humans, males, or females), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, and the like. In certain embodiments, the subject is a primate. In a preferred embodiment, the subject is a human. The terms "subject" and "patient" are used interchangeably when referring to a human.

As used herein, a subject is "in need of" a treatment if the subject would benefit biologically, medically or in quality of life from such treatment.

As used herein, the phrase "patient population" is used to mean a group of patients.

The term "comprising" encompasses "including" as well as "consisting of … …," e.g., a composition "comprising" X may consist of X alone or may include other materials, such as X + Y.

The term "about" in relation to the value x means, for example, +/-10%. The term "about" applies to each number in a series when used before a numerical range or list of numbers, for example, the phrase "about 1-5" should be interpreted as "about 1-about 5", or for example, the phrase "about 1, 2, 3, 4" should be interpreted as "about 1, about 2, about 3, about 4, etc.

The term "treatment" is defined herein as the application or administration of a compound according to the present disclosure (a compound having formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the compound) to a subject or to an isolated tissue or cell line from a subject, wherein the subject has a particular disease (e.g., SjS), has symptoms associated with the disease (e.g., SjS) or a predisposition to develop (if applicable) to the disease (e.g., SjS), wherein the purpose of treatment is to cure (if applicable) the disease, delay the onset of the disease, reduce its severity, alleviate, ameliorate one or more symptoms of the disease, ameliorate the disease, reduce or ameliorate any symptoms associated with the disease or the predisposition to develop to the disease. The term "treating" includes treating a patient suspected of having a disease as well as a patient who is ill or has been diagnosed with a disease or medical condition, and includes inhibiting clinical relapse.

As used herein, "selecting" and "selected" with respect to a patient is used to mean that a particular patient is specifically selected from a larger group of patients based on (due to) the particular patient having predetermined criteria. Similarly, "selective treatment" refers to providing treatment to a patient suffering from a particular disease, wherein the patient is specifically selected from a larger group of patients based on the particular patient having predetermined criteria. Similarly, "selectively administering" refers to administering a drug to a patient specifically selected from a larger group of patients based on (due to) the particular patient having predetermined criteria. By "selecting," "selective treating," and "selectively administering," it is meant delivering personalized therapy to a patient based on the patient's personal medical history (e.g., previous therapeutic intervention, such as previous treatment with a biological agent), biological characteristics (e.g., a particular genetic marker), and/or performance (e.g., not meeting a particular diagnostic criteria), rather than delivering a standard treatment regimen based solely on the patient's membership in a larger group. With reference to a method of treatment as used herein, selection does not refer to an incidental treatment of a patient having a particular criteria, but rather refers to an intentional selection of a patient for treatment based on a patient having a particular criteria. Thus, selective treatment/administration is different from standard treatment/administration that delivers a particular drug to all patients with a particular disease, regardless of their personal medical history, disease manifestation, and/or biological characteristics. In some embodiments, the patient is selected for treatment based on being afflicted with SjS.

The specific implementation mode is as follows:

sjogren's syndrome and effectiveness of treatment according to the invention

The disclosed BTK inhibitors, i.e., compounds having formula (I), or pharmaceutically acceptable salts thereof, can be used in vitro, ex vivo, or incorporated into pharmaceutical compositions and administered in vivo to treat SjS patients (e.g., human patients).

The effectiveness of treatment of sjogren's syndrome can be assessed using various known methods and tools that can measure the state of sjogren's syndrome and/or the clinical response of sjogren's syndrome. Some examples include, for example, EULAR Sjogren's Syndrome Disease Activity Index (ESSDAI), physician comprehensive assessment scale (PhGA), EULAR Sjogren's Syndrome Patient Reporting Index (ESSPRI), chronic disease treatment function assessment-fatigue scale (FACIT-fatigue), and EQ 5D.

Therapeutic effect

Clinical efficacy measures associated with primary and secondary objectives are summarized below.

EULAR Sjogren Syndrome Disease Activity Index (ESSDAI)

ESSDAI is a validated disease outcome measure against Sjogren's syndrome and is applied to study subjects (Seror R et al (2015) differentiation of EULAR primary)Verification of syndrome disease activity (ESSDAI) and patient indices (ESSPRI) [ EULAR Primary Sjogren's syndrome disease Activity (ESSDAI) and patient indices (ESSPRI)]Ann. Rheum. Dis. [ year of rheumatic diseases]Pages 859-66). The instrument contains 12 organ-specific domains that contribute to disease activity. For each domain, the characteristics of disease activity were scored at 3 or 4 levels according to their severity. These scores are then summed in a weighted fashion over the 12 domains to provide a total score. The fields (weights) are as follows: constitution (3), lymph node lesions (4), glands (2), joints (2), skin (3), lungs (5), kidneys (5), muscles (6), PNS (5), CNS (5), hematology (2) and biology (1). The highest possible score was 123.

In our study, to calculate ESSDAI, all 12 organ domains must be evaluated individually at each scheduled time point (from screening visit to study end). The domain evaluations are entered into a table (provided by the central vendor) and the ESSDAI scores are calculated by the software.

For assessments that are not listed as mandatory tests in the protocol, but may be required to estimate ESSDAI, including radiography, High Resolution Computed Tomography (HRCT), pulmonary function tests (DLCO, FVC), estimated glomerular filtration rate (eGFR), Electromyography (EMG), muscle (or any other) biopsy, a researcher decides whether to evaluate based on the patient's signs and symptoms to provide the correct ESSDAI reading. EULAR sjogren's syndrome disease index (ESSDAI), domain and entry definitions and weights are summarized in table 1:

doctor comprehensive assessment scale (PhGA)

The investigator rated the disease activity of the patient from "no disease activity" (0) to "maximum disease activity" (100) by a 100mm VAS using the physician's full assessment scale.

To enhance the objectivity, the physician is unaware of the outcome assessment reported by a particular patient when making his own assessment of that particular patient. Therefore, the assessment must be made before looking at a comprehensive assessment of the patient's overall disease activity score.

EULAR Sjogren Syndrome Patient Reporting Index (ESSPRI)

ESSPRI is a given disease outcome measure for Sjogren's syndrome (Seror R et al (2011) EULAR)Syndrome Patient Reported Index(ESSPRI):development of a consensus patient index for primarysyndrome [ EULAR sjogren's syndrome patient reporting index (espfri): establishment of consensus index of patients with primary sjogren's syndrome]Ann. Rheum. Dis. [ year of rheumatic diseases]Pages 968-72). It consists of three domains, dryness, pain and fatigue. Subjects can assess the severity of symptoms they experience on a single 0-10 numerical scale for each of the three domains. The ESSPRI score is defined as the average of the scores from three scales: (dry + pain + fatigue)/3.

FACIT-fatigue

The chronic disease treatment function assessment-fatigue scale (FACIT-F v4) is a short, easy to operate 13-item tool that can be used to measure the degree of fatigue in an individual during the past week of daily activity. The degree of fatigue is measured on a 5 point litter scale (0: none, 1: one, 2: slight, 3: equivalent, 4: extraordinary) (Webster K et al (2003) The Functional Assessment of Chronic Illness Therapy (FACIT) Measurement System: properties, applications and interpretation [ Chronic Life Therapy Functional Assessment (FACIT) Measurement System: properties, application and interpretation ]. Health care Life issues [ Health and quality of Life results ] page 79).

EQ5D

EQ-5D is a standardized tool for measuring health-related quality of life.

EQ-5D consists of a description system and EQ VAS scale.

The description system includes five dimensions: mobility, self-care, daily activities, pain/discomfort and anxiety/depression. It can be used as a quantitative measure reflecting the health outcome of the patient's own judgement. The scores in these five dimensions may be presented as health conditions, or may be converted to a single aggregate index number (utility) reflecting a preference over other health conditions.

EQ VAS records the patient's self-assessed health condition on a vertical visual analog scale, where 0 represents the ' worst imaginable health state ' and 100 represents the ' best imaginable health state '.

Appropriateness of efficacy assessment

Efficacy measures in this study were based primarily on ESSDAI (EULAR SS disease Activity index) which measures organ-specific disease criteria, and ESSPRI (European Union of antirheumatics [ EULAR)]Sjogren's syndrome [ SS]Patient reporting index). Both of these tools are widely accepted and validated, being the gold standard measure of systemic and symptomatic performance of SjS, respectively. ESSDAI is a systemic disease activity index, dividing disease activity into 3-4 levels in each of 12 differentially weighted domains (biological, hematological, joint, glandular, skin, constitutional, lymphadenopathy, kidney, lung, PNS, CNS, and muscle). The composite weighted score provides an accurate assessment of disease activity and has good sensitivity to changes, as has been validated in multiple cohort studies (Seror R et al (2015) validity of EULAR primary)Verification of syndrome disease activity (ESSDAI) and patient indices (ESSPRI) [ EULAR Primary Sjogren's syndrome disease Activity (ESSDAI) and patient indices (ESSPRI)]Ann. Rheum. Dis. [ year of rheumatic diseases]Pages 859-66). On the other hand, the ESSPRI tool is a composite score of the dry, limb pain and fatigue symptoms assessed on a 0-10 visual analog scale during the first 2 weeks reported by the patient (Seror R et al (2011) EULAR)′sSyndrome Patient Reported Index(ESSPRI):development of a consensus patient index for primarysyndrome [ EULAR sjogren's syndrome patient reporting index (espfri): establishment of consensus index of patients with primary sjogren's syndrome]Ann. Rheum. Dis. [ year of rheumatic diseases]Pages 968-72). The patient reported scores were less sensitive to changes in disease activity, while the ESSPRI was reported to have significantly better sensitivity among the available tools. A recent prospective study reported a poor correlation between whole body and patient scoresThis indicates that these two indices evaluate the complementary components of disease activity, thus emphasizing the importance of evaluating two parameters to accurately assess disease activity and its changes (Seror R et al (2015) differentiation of EULAR primary)Verification of syndrome disease activity (ESSDAI) and patient indices (ESSPRI) [ EULAR Primary Sjogren's syndrome disease Activity (ESSDAI) and patient indices (ESSPRI)]Ann. Rheum. Dis. [ year of rheumatic diseases]Pages 859-66).

Pharmaceutical composition

BTK inhibitors, i.e., compounds having formula (I), or pharmaceutically acceptable salts thereof, can be used as pharmaceutical compositions when combined with a pharmaceutically acceptable carrier. In addition to the compound having formula (I), such compositions can also comprise carriers, various diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials known in the art. The characteristics of the vector depend on the route of administration. The pharmaceutical compositions for use in the disclosed methods may also contain other therapeutic agents for treating specific targeted disorders. For example, the pharmaceutical composition may also include an anti-inflammatory or anti-pruritic agent. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect with, or minimize side effects caused by, the compounds of formula (I) described herein. In preferred embodiments, the pharmaceutical composition for use in the disclosed methods comprises a dose of 10mg, 20mg, 25mg, 50mg, or about 100mg of the compound having formula (I).

Suitable compositions for oral administration include an effective amount of a compound of the invention in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of: sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients are, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. The tablets are uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. Formulations for oral use may be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.

Pharmaceutical compositions for use in the disclosed methods can be prepared in a conventional manner. In one embodiment, the pharmaceutical composition is provided for oral administration. For example, the pharmaceutical composition is a tablet or gelatin capsule comprising the active ingredient and:

a) diluents, such as lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine;

b) lubricants, for example silica, talc, stearic acid, magnesium or calcium salts thereof and/or polyethylene glycol; in the case of tablets, further comprising

c) Binders, such as magnesium aluminum silicate, starch paste, gelatin, gum tragacanth, methyl cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone; if desired

d) Disintegrating agents, such as starch, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or

e) Adsorbents, colorants, flavors, and sweeteners.

Tablets may be film coated or enteric coated according to methods known in the art.

Combining:

in practicing some of the therapeutic methods or uses of the present disclosure, a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered to a patient, such as a mammal (e.g., a human). While it is understood that the disclosed methods provide treatment for sjogren's syndrome patients using a compound having formula (I) or a pharmaceutically acceptable salt thereof, the therapy is not necessarily monotherapy. Indeed, if a patient is selected for treatment with a compound having formula (I), the compound having formula (I) or a pharmaceutically acceptable salt thereof may be administered according to the methods of the present disclosure alone or in combination with other agents and therapies for treating sjogren's syndrome patients, for example in combination with at least one additional sjogren's syndrome agent. When co-administered with one or more additional SjS agents, the compound having formula (I) or a pharmaceutically acceptable salt thereof can be administered simultaneously or sequentially with the other agents. If administered sequentially, the attending physician decides the appropriate order of administration of the compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with other agents and the appropriate dosage for co-delivery.

During treatment SjS, various therapies can be beneficially combined with the disclosed compounds having formula (I) or pharmaceutically acceptable salts thereof. Such therapies include steroids (corticosteroids, such as prednisone or equivalent); DMARDS, such as hydroxychloroquine (plaquinil), methotrexate (Trexall), sulfasalazine (Azulfidine), minocycline (Minocin), or leflunomide (Arava)); or a B cell depleting drug, such as rituximab.

The skilled artisan will be able to identify an appropriate dose of the SjS agents described above for co-delivery with the disclosed compounds of formula (I) or pharmaceutically acceptable salts thereof.

Kit of the invention

The present disclosure also encompasses kits for treating SjS. Such kits comprise a BTK inhibitor, such as N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutical composition thereof. Additionally, such kits may comprise instructions for use.

In one embodiment, the kit comprises two or more separate pharmaceutical compositions, at least one of which comprises a compound having formula (I) or a pharmaceutically acceptable salt thereof. In one embodiment, the kit comprises a device for separately retaining the compositions, such as a container, a separate bottle, or a separate foil packet. Examples of such kits are blister packs, as typically used for packaging tablets, capsules and the like.

The kits of the invention can be used for administering different dosage forms (e.g., oral and parenteral), for administering separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To aid compliance, the kits of the invention typically contain instructions for administration.

In the combination therapies of the present invention, the compound having formula (I) or a pharmaceutically acceptable salt thereof and the other SjS agent (as defined herein) may be produced and/or formulated by the same or different manufacturers. Further, the compound having formula (I) or a pharmaceutically acceptable salt thereof and another SjS agent may be combined in a combination therapy: (i) prior to dispensing the combination product to a physician (e.g., in the case of a kit comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof and another SjS medicament); (ii) by the physician himself (or under the direction of the physician) shortly before administration; (iii) in the patient himself, for example during sequential administration of a compound of formula (I) or a pharmaceutically acceptable salt thereof and the other SjS agent.

Other embodiments

The compound having formula (I) or a pharmaceutically acceptable salt thereof is conveniently administered to the patient (preferably orally) at a dose of from about 10mg to about 200mg per day.

The compound having formula (I) or a pharmaceutically acceptable salt thereof is conveniently administered to the patient (preferably orally) in a daily dose of from about 10mg to about 200mg per day.

In some embodiments, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered in a daily dose of about 10mg to about 100 mg.

In other embodiments, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered in a daily dose of about 100 mg.

In other embodiments, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered in a daily dose of about 50 mg.

In other embodiments, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered at a daily dose of about 35 mg.

In other embodiments, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered at a daily dose of about 25 mg.

In other embodiments, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered at a daily dose of about 20 mg.

In one embodiment, a compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered once daily at a dose of about 10mg, about 35mg, about 50mg, or about 100 mg.

In another embodiment, a compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered twice daily at a dose of about 10mg, about 25mg, about 50mg, or about 100 mg.

It is to be understood that certain patients may require dose escalation, for example patients exhibiting an inadequate response to sjogren's syndrome (e.g., as measured by any of the sjogren's syndrome scoring systems disclosed herein. it is also to be understood that certain patients, for example patients exhibiting an adverse event or adverse response to treatment with a compound of formula (I) or a pharmaceutically acceptable salt thereof, may also require a reduced dose.

The time of administration is typically measured from the day (also referred to as "baseline") of the first dose of the compound having formula (I) or a pharmaceutically acceptable salt thereof. The time of administration is typically measured from the day (also referred to as "baseline") of the first dose of the compound having formula (I) or a pharmaceutically acceptable salt thereof.

However, healthcare providers often use different naming conventions to determine the dosing schedule. For clarity, the first day of administration is referred to as day 1, as disclosed herein. However, those skilled in the art will appreciate that the nomenclature is merely used uniformly and should not be construed as limiting, i.e., daily administration is to provide a daily dose of a compound of formula (I) or a pharmaceutically acceptable salt thereof, and a physician may refer to a particular day as "day 0" or "day 1".

Disclosed herein are methods of treating sjogren's syndrome (SjS) comprising administering to a patient in need thereof a compound having formula (I) or a pharmaceutically acceptable salt thereof, wherein the dose is from about 10mg to about 200 mg.

Also disclosed herein are methods of treating sjogren's syndrome (SjS) comprising administering to a patient in need thereof a compound having formula (I) or a pharmaceutically acceptable salt thereof, wherein the daily dose is from about 10mg to about 200 mg.

Also disclosed herein is a compound having formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of SjS, wherein the daily dose of the compound is from about 10mg to about 200 mg.

In one embodiment of the disclosed methods, uses and kits, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered in a daily dose of about 10mg to about 100 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered in a daily dose of about 10mg, about 20mg, about 25mg, about 35mg, about 50mg, about 100mg or about 200 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered in a daily dose of about 100 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I) or a pharmaceutically acceptable salt thereof is administered in a daily dose of about 50 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I) or a pharmaceutically acceptable salt thereof is administered in a daily dose of about 35 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I) or a pharmaceutically acceptable salt thereof is administered in a daily dose of about 25 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I) or a pharmaceutically acceptable salt thereof is administered in a daily dose of about 20 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered once daily at a dose of about 10mg, about 35mg, about 50mg, or about 100 mg.

In another embodiment of the disclosed methods, uses and kits, the compound having formula (I), or a pharmaceutically acceptable salt thereof, is administered twice daily at a dose of about 10mg, about 25mg, about 50mg, or about 100 mg.

In another embodiment of the disclosed methods, uses and kits, the patient has moderate to severe SjS. A patient with moderate to severe SjS is defined as having an ESSDAI score (based on the weighted scores shown in table 1) of > 5 (i.e., at least 5 points) from 8 defined domains (biological, hematological, joint, skin, glandular, lymph node lesion, kidney, physical) and an ESSPRI score of at least 5 points prior to treatment with a compound having formula (I) or a pharmaceutically acceptable salt thereof.

In another embodiment of the disclosed method, use and kit, the patient is an adult.

In another embodiment of the disclosed methods, uses and kits, the patient achieves a change in at least one of the patient's and/or physician's reported results (i.e., ESSPRI, FACIT-F, EQ-5D, PhGA) at week 12 or week 24 of treatment as compared to baseline.

In another embodiment of the disclosed methods, uses and kits, the patient achieves a change in the espri score from baseline at week 12 or week 24 of treatment.

In another embodiment of the disclosed methods, uses and kits, the patient achieves a decrease in the espri score at week 12 or 24 of treatment.

In another embodiment of the disclosed method, use and kit, the patient achieves at least one, preferably at least 2, minutes of reduction in the espri score at week 12 or 24 of treatment.

In another embodiment of the disclosed methods, uses and kits, the patient achieves a decrease in the espri score at week 12 or 24 of treatment. In another embodiment of the disclosed methods, uses and kits, the patient achieves at least a 15%, at least a 25%, at least a 35%, at least a 50% or at least a 60% reduction in the espri score at week 12 or 24 of treatment. The reduction in the ESSPRI score is calculated as follows:

in another embodiment of the disclosed methods, uses and kits, the patient achieves a decrease in ESSDAI score at week 12 or 24 of treatment.

In yet another embodiment of the disclosed methods, uses and kits, the patient achieves a reduction in ESSDAI score of at least 3 points at week 12 or 24 of treatment.

In another embodiment of the disclosed methods, uses and kits, the patient achieves a change in the ESSDAI score from baseline at week 12 or 24 of treatment.

In preferred embodiments of the disclosed methods, uses and kits, the patient is an adult. In some embodiments of the disclosed methods, uses and kits, the patient is a juvenile.

Examples are further illustrated

1. A method of treating sjogren's syndrome (SjS), comprising administering to a subject in need thereof a daily dose of about 10mg to about 200mg of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof.

2. The method of embodiment 1, wherein the daily dose of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is from about 10mg to about 100 mg.

3. The method of embodiment 1 wherein the daily dose of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is about 100 mg.

4. The method of embodiment 1 wherein the daily dose of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is about 50 mg.

5. The method of embodiment 1, wherein the daily dose of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is about 35 mg.

5. The method of embodiment 1 wherein the daily dose of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is about 25 mg.

7. The method of embodiment 1, wherein the daily dose of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is about 20 mg.

8. The method of embodiment 1, wherein N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is administered at a dose of about 10mg, about 35mg, about 50mg, or about 100mg once daily.

9. The method of embodiment 1, wherein N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereof is administered at a dose of about 10mg, about 25mg, about 50mg, or about 100mg twice daily.

10. The method of any one of the above embodiments, wherein the subject has moderate to severe SjS.

11. The method according to any one of embodiments 1-10, wherein the subject is selected according to at least one of the following criteria:

a) (ii) the ESSPRI score of the subject is ≧ 5 prior to treatment with the compound having formula (I) or a pharmaceutically acceptable salt thereof;

b) the subject's weight-based score of ESSDAI ≧ 5 from 8 specific domains of biology, hematology, joint, skin, gland, lymph node pathology, kidney, and constitution prior to treatment with the compound having formula (I) or a pharmaceutically acceptable salt thereof.

12. The method of any one of the above embodiments, wherein the subject is an adult.

13. The method according to any one of the preceding embodiments, wherein the subject achieves at least one of the following at week 12 or 24 of treatment:

a) a decrease in the ESSPRI score; and/or

b) A decrease in the ESSDAI score.

14. The method of any one of the above embodiments, wherein the subject achieves a sustained response as measured by espri or ESSDAI by week 5 after completion of the treatment.

15. The method of any one of the above embodiments, wherein N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide, or a pharmaceutically acceptable salt thereof, is formulated in a pharmaceutical formulation, wherein the pharmaceutical formulation further comprises a pharmaceutically acceptable carrier.

16. The method of any one of embodiments 1-15, wherein T of N- (3- (6-amino-5- (2- (N-methacrylamido) ethoxy) pyrimidin-4-yl) -5-fluoro-2-methylphenyl) -4-cyclopropyl-2-fluorobenzamide or a pharmaceutically acceptable salt thereofmaxFrom about 0.5 to about 3 hours.

The details of one or more embodiments of the disclosure are set forth in the accompanying description above. Preferred methods and materials are now described, but any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure. Other features, objects, and advantages of the disclosure will be apparent from the description and from the claims. In this specification and the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents and publications cited in this specification are incorporated by reference. The following examples are provided in order to more fully illustrate the preferred embodiments of the present disclosure. These examples should in no way be construed as limiting the scope of the disclosed subject matter as defined by the appended claims.

Abbreviations

AE adverse events

Area under AUC curve

Area under the plasma (or serum or blood) concentration-time curve (mass x time/volume) from time zero to infinity of AUCinf

Area under the plasma (or serum or blood) concentration-time curve (mass x time/volume) from time zero to time of the last quantifiable concentration of AUClast

Area under the plasma (or serum or blood) concentration-time curve (mass x time/volume) of AUC τ from time zero to the end of dosing interval τ

BCR B cell receptors

Bid or b.i.d. twice daily (Latin: "bis in die")

BMI body mass index

BTK Bruton tyrosine kinase

CBC complete blood cell count

cm

Apparent systemic (or systemic) clearance (mass/volume) of CL/F plasma (or serum or blood)

CNS central nervous system

Coefficient of variation of CV

Antirheumatic DMARD-ameliorating agents

ECG electrocardiogram

eGFR estimation of glomerular filtration rate

ELISA enzyme-linked immunosorbent assay

EMG electromyogram

EQ-5D EuroQual 5 dimension (Standard tool for measuring quality of Life related to health)

ESSDAI EULAR Sjogren syndrome disease activity index

ESSPRI EULAR Sjogren syndrome patient reporting index

EULAR European antirheumatic Union

Assessment of therapeutic function of FACIT-F Chronic disease-fatigue

FIH first human body

h hours

HRCT high resolution computed tomography

i.v. intravenous

Analysis in phase IA

INR International normalized ratio

kg kilogram

LC-MS/MS liquid chromatography/mass spectrometry-mass spectrometry

mAb monoclonal antibodies

MCP-Mod multiple comparison program-modeling

MMRM mixed effect model repetition measurement

MRT mean residence time

NOAC novel oral anticoagulant

NSAID non-steroidal anti-inflammatory drugs

PD pharmacodynamics

PhGA physician comprehensive assessment scale

PK pharmacokinetics

PNS peripheral nervous system

PT prothrombin time

PTT partial thromboplastin time

Once daily Qd or q.d. (Latin words "quaque die")

QTcF QT interval corrected by Fridericia formula

Racc drug accumulation Rate

SAE Severe adverse events

SjS Sjogren's syndrome

SOM field operation manual

SPT skin prick test

SS security suite

TEC tyrosine protein kinase

Apparent volume of distribution (volume) at terminal elimination stage after Vz/F administration

Example 1: preclinical study

Example 1 a: BTK occupancy and preclinical PK/PD relationships

The in vivo PD effect of irreversible BTK inhibitors like compound (I) depends on the extent and duration of covalent occupancy of BTK by the inhibitor. BTK occupancy after treatment with a compound having formula (I) (also referred to as compound (I)) was measured in an ex vivo immunoassay. The proportion of unoccupied BTK protein was determined after incubation with covalently biotinylated BTK probe in vitro, since compound (I) and probe bound BTK in a mutually exclusive manner. The unoccupied BTK as well as the total BTK relative protein level were determined in lysates of selected tissues and normalized to the total BTK protein level in the same sample.

In female rats, a single oral dose of 3mg/kg of compound (I) resulted in complete occupancy of spleen BTK, whereas a dose of 1mg/kg resulted in 76% -81% occupancy, whereas a single dose of 0.3mg/kg only reached a partial occupancy of 30%. The occupancy of BTK in blood reached a level consistent with that observed in the spleen. From experimental data, it is clear that a brief systemic exposure of compound (I) at low oral doses of 1-3mg/kg is sufficient to achieve complete occupancy of BTK in multiple tissues. After a 1mg/kg dose, blood exposure of compound (I) reached 49.1nM at 0.5 hours post-dose and 5.6nM at 5 hours post-dose. This extremely low and short systemic exposure is consistent with the PK/PD model typical of irreversible inhibitors.

The duration of BTK occupancy of spleen, blood, lymph nodes and lung in rats and mice was determined after a single oral dose of compound (I). In rats, the half-life occupied by BTK in blood is long, about 87 hours. The estimated half-life of BTK occupancy in rat spleen is significantly shorter than in blood, only about 5 hours. The different turnover rates probably reflect the fact that BTK-expressing B cells and monocytes are quiescent and relatively metabolically inactive in peripheral blood compared to spleen. Longer durations of BTK occupancy in blood have been previously reported (Advani et al 2013, J Clin Onc [ J. Clin Oncs.; 31(1): 88-94). All other tissues analyzed (lung and lymph nodes) showed similar BTK turnover and occupancy half-life as spleen.

Since the level of BTK-expressing cells in the skin is too low to make occupancy measurements, the duration of PD effects in the skin after a single dose was evaluated in a passive arabidopsis (RPA) model of reversal of mast cell Fc γ RIII-mediated inflammation. In this model, inhibition of skin swelling was greatest when compound (I) was administered 2 hours before initiating the ather reaction. This effect gradually decreased and reached baseline when the atse response was triggered 45 hours or later after compound (I) administration. This indicates that BTK occupancy in skin shows a similar time course as e.g. spleen, lung and lymph nodes.

In these preclinical pharmacological studies, BTK occupancy and the respective pharmacological readings show strong correlation. Thus, BTK occupancy is a PD biomarker suitable for clinical studies, and is therefore used in phase 1 clinical studies.

Example 2: phase 1 clinical trial

A first human study was conducted to evaluate the safety and tolerability, Pharmacokinetics (PK) and Pharmacodynamics (PD) of oral administration of single or multiple doses of compound (I) once daily (qd) or twice daily (bid) in healthy and atopic constitutional volunteers to support further clinical development of compound (I) in autoimmune diseases. This study also explored the effect of food intake.

The first human study was conducted in up to 168 Healthy Volunteers (HV), 64 of which (in parts 2 and 4) had asymptomatic atopic constitution.

Section 1 is a double-blind (blind to subjects and investigators, not to sponsor), placebo-controlled single-increment dose (SAD) escalation study (N-80) of 10 cohorts

Part 2 is a double-blind (blind to subjects and investigators, not to the applicant), placebo-controlled Multiple Ascending Dose (MAD) (13 doses in 12 days) study with once-daily dosing (N ═ 48) in 6 groups of healthy volunteers with asymptomatic atopy constitution

Section 3 is a single dose open label cross-feed effect study in 12 HV

Part 4 is a double-blind (blind to subjects and investigators, not to sponsor), placebo-controlled multiple dose (12 day 25 dose) study with twice daily dosing in 2 groups of asymptomatic atopic healthy volunteers (N ═ 16)

The SAD section (section 1) had ten dose levels and the MAD section (sections 2 and 4) consisted of eight dose levels (6 cohorts in section 2 used once daily dosing and 2 cohorts in section 4 used twice daily dosing). Eight subjects were randomized into groups and received compound (I) or matched placebo at a ratio of 6:2 (active: placebo) in the SAD and MAD sections, respectively. The dose in the SAD section, up to about 4 times the estimated Pharmacologically Active Dose (PAD), should be evaluated prior to the beginning of the MAD section of the study, provided that no safety signal was present in the SAD section before this time. The total daily dose of compound (I) used in sections 2 (madqd regimen) and 4 (multiple dose bid regimen) did not exceed the highest SAD dose level at hand. Further, the total daily dose of part 4 does not exceed the total daily dose of part 2.

In section 1 (SAD), the first sentinel dosing was performed at each dose level as described below. The first two subjects were dosed on the first day (one with active drug and one with placebo). After an observation period of 48 hours, the remaining 6 subjects of the group (5 with active drug, 1 with placebo) were dosed.

Standard safety monitoring was used in all study sections. A specific assessment of potential skin abrasion events is included. All vital signs, physical examination and subject history, ECG, adverse events and laboratory safety parameters (blood chemistry, hematology and urinalysis) for up to 96 hours after the last dose, and PK data for previous dose groups (if any) for 48 hours after the last dose, should be reviewed blindly before dose escalation for each cohort. After completion of each dose level, a summary safety report of reported adverse events, clinical safety laboratory parameters, QTc and heart rate is provided.

In sections 1, 2 and 4, each subject participated in a 28-day screening period (days-29 to-2), baseline period, treatment period and follow-up period including end-of-study assessment.

In part 1, subjects entered the study center on day-2 or day-1 for baseline safety assessment and confirmation of eligibility. Eligible subjects received a single dose of compound (I) or placebo under fasting conditions on day 1. They lived at the indicated sites from day-1 to day-5 morning (96 hours after the last drug administration).

In parts 2 and 4, subjects were admitted to the hospital on days-2 or-1 for baseline safety assessment and confirmation of eligibility. Eligible subjects received a first dose of compound (I) under fasting conditions on day 1 and continued to take study medication under fasting conditions until day 12 (including day 12). Subjects lived at the indicated sites starting on day-2 or-1 until day 16 morning (equal to 96 hours after receiving the last dose of compound (I)). In parts 2 and 4, the study drug was administered once daily and twice daily, respectively (detailed information can be found in the evaluation schedule).

Section 3 is an open label, randomized, two-way crossover, single dose study to assess food effect. In part 3, each subject participated in a 28-day screening period (-29 to-2 days), 2 baseline periods (day-1), and 2 treatment periods, each consisting of a single dose administration on day 1 and subsequent safety and PK assessments until day 5. Treatment period 2 included a follow-up visit and study end evaluation on day 22 and day 40, respectively. The two treatment periods are separated by an elution period of at least 18 days (+/-1 day).

One or more primary targets

And (3) inclusion standard:

1. healthy male and female subjects between the ages of 18 and 65 (inclusive) were determined to be in good condition by past medical history, physical examination, vital signs, electrocardiogram and laboratory examination at screening. Healthy subjects with atopic constitution participated in either part 2 or part 4, according to the qualifications of these particular study groups. Atopic healthy volunteers must be positive for skin prick tests with known allergens at screening (atopic constitution), but clinically asymptomatic, without any systemic drug treatment.

2. The subject is required to weigh at least 50kg and have a Body Mass Index (BMI) in the range of 18-30kg/m2, inclusive. BMI ═ weight (kg)/[ height (m)]2

3. At screening and first baseline, the subjects were assessed in a sitting position after sitting still for at least 3 minutes and again (if needed) after standing for 3 minutes (body temperature, systolic and diastolic blood pressure and pulse rate). The sitting vital signs must be within the following ranges (inclusive):

oral body temperature between 35.0 ℃ and 37.5 ℃

A systolic pressure of 90 to 139mm Hg

Diastolic pressure of 50 to 89mm Hg

Pulse rate of 50 to 90bpm

Key exclusion criteria

1. There was a history of allergy to any of the study drugs or similar chemical classes of drugs.

2. A history of clinically significant ECG abnormalities, or any of the following ECG abnormalities prior to screening and/or treatment:

PR interval >200 ms

QRS complex >120 ms

QTcF >450 ms (Male)

QTcF >460 ms (female)

3. Hemoglobin levels at screening or first baseline were below 12.0 g/dL.

4. Platelet count at screening or first baseline out of normal range (below 150X 10)9/L or greater than 450x 109)。

5. Any clinically significant abnormality in any standard coagulation test, including Prothrombin Time (PT), Partial Thromboplastin Time (PTT) or International Normalized Ratio (INR), at screening and/or baseline.

6. There is a history of or an increased risk of a thromboembolic or thromboembolic event.

Treatment of administration

Section 1 (SAD)

Subjects were assigned to one of the following 10 groups. In each cohort, 8 subjects were randomly assigned to receive compound (I) or matched placebo at an overall 6:2 ratio. The first sub-group was randomly assigned to compound (I) for one subject and a matching placebo for another at a ratio of 1: 1. The remaining 6 subjects/cohort administered after the 48 hour observation period of the initially administered 2 subjects were randomly assigned at a ratio of 5: 1.

Group 1: single oral dose 0.5mg of Compound (I) or matching placebo

Group 2: single oral dose 1.5mg of Compound (I) or matching placebo

Group 3: single oral dose 5mg of Compound (I) or matched placebo

Group 4: single oral dose 15mg of Compound (I) or matched placebo

Group 5: single oral dose 30mg of Compound (I) or matched placebo

Group 6: single oral dose 60mg of Compound (I) or matched placebo

Group 7: single oral dose 100mg of Compound (I) or matched placebo

Group 8: single oral dose 200mg of Compound (I) or matched placebo

Group 9: single oral dose 400mg of Compound (I) or matching placebo

Group 10: single oral dose 600mg of Compound (I) or matching placebo

Part 2 (MAD, qd scheme)

Subjects were assigned to one of the following 6 groups. In each cohort, 8 subjects received compound (I) or matched placebo at a ratio of 6:2 randomly assigned.

Group 1: multiple oral doses of 10mg of Compound (I) or matched placebo

Group 2: multiple oral doses of 25mg of Compound (I) or matched placebo

Group 3: multiple oral doses of 50mg of Compound (I) or matched placebo

Group 4: multiple oral doses of 100mg of Compound (I) or matched placebo

Group 5: multiple oral doses of 400mg of Compound (I) or matched placebo

Group 6: multiple oral doses up to 600mg of Compound (I) or matched placebo

Section 3 (food effect)

Subjects were randomly assigned to one of 2 treatment orders at a 1:1 ratio:

section 4 (MAD, bid scheme)

The subjects were assigned to one of the following groups. In each cohort, 8 subjects received compound (I) or matched placebo at a ratio of 6:2 randomly assigned.

Group 1: multiple oral doses of 100mg of Compound (I) or matched placebo in the bid regimen

Group 2: multiple oral doses of 200mg of Compound (I) or matched placebo in the bid regimen

Pharmacokinetic data

The biological analysis method comprises the following steps:

pharmacokinetic samples were obtained from blood at all dose levels in all subjects and evaluated. No samples from placebo subjects were analyzed. PK assessment samples from subjects were collected at defined time points in the study. The concentration of compound (I) in blood was determined by a validated LC-MS/MS method.

Single ascending dose 0.5mg-600mg pharmacokinetics:

the mean plasma concentration-time course of compound (I) after a single ascending dose is shown in figure 1.

At all doses, compound (I) was rapidly absorbed over time, reaching a Cmax of about 1-1.5 hours. In most subjects, the absorption phase is characterized by a single distinct absorption peak. Drug treatment showed a bi-exponential decline. Most of the drug was eliminated during the initial dispensing phase, indicating that a significant amount of drug clearance may occur before systemic tissue balance is reached. The apparent terminal elimination phase is not reached until 12 hours post-dose and is only measurable in subjects receiving doses of 100mg and above. The measurable terminal half-life is 4 hours (100mg) to 18 hours (600mg), resulting in an average residence time in circulation (MRT) of 1 hour to 5 hours (MRT ≈ T1/2/ln 2). The partitioning phase showed a major dose dependence of about 1h, T1/2. In the SAD group, the geometric mean of oral blood clearance after single dose administration (CL/F) ranged from 250 to 506L/h, estimated to be 383L/h in all groups.

Multiple oral dose pharmacokinetics

Figure 2 shows the mean plasma concentration of compound (I) over time after multiple escalating doses of 10mg to 400 mg.

In each group, the geometric mean apparent clearance at steady state (CLss/F, day 12 MAD, q.d.) after oral administration was between 246 and 414L/h. Generally, lower clearance was observed at steady state compared to day 1, but at doses of 100mg and above, this difference almost disappeared (tables 2-1 (day 1) and 2-2 (day 12)). The reason for this behavior may be that covalent target (BTK) binding contributes to the initial clearance of compound (I). This effect was most pronounced on day 1, as the residual target occupancy in the trough decreased the partial contribution of target binding clearance (CLss/F) over consecutive days. Naturally, as target occupancy in the trough approaches completion, the difference decreases with increasing dose. Thus, drug exposure (AUC, Cmax) was higher at day 12 than at day 1, ranging from 5 (low dose) to 1.2 (high dose) doses as shown by the drug accumulation rate (Racc) within the subject, and AUC was generally higher than Cmax, confirming that effects on systemic clearance may be implicated.

TABLE 2-1: summary of PK parameters for compound (I) at multiple ascending doses of 10-600mg q.d. administration

Tables 2 to 2:

statistics as mean. + -. SD (CV%)

Median (Min-Max) [ n ]

CV% (% of variation) (% of SD)/average value) (% of 100

For Tmax and T1/2, only the median (Min-Max) [ n ] is shown

Typically, the blood concentration 24 hours after the last administration is usually below 1ng/ml, except for some subjects with doses of 100mg or higher, which indicates that compound (I) is almost completely eluted in two consecutive doses. The latter also indicates that steady state is reached within a few doses.

B.i.d. dosing was also studied due to the higher turnover rate of BTK in tissues. Fig. 3 shows the mean plasma concentration time profiles obtained after multiple ascending doses of 100mg and 200mg twice daily. Consistent with the results of the other groups, a rapid absorption of the dose after the b.i.d. regimen was observed, Tmax of about 1 h. The observed cumulative factor for AUC (Racc) reached 1.5(100mg) and 2.0(200mg), and the cumulative factor for Cmax reached about 1.65 (two doses). A dose proportional increase in AUC τ was observed at day 12, while only a slight increase in Cmax was found (1.33-fold). In summary, b.i.d administration of compound (I) provides an option to address faster target re-synthesis in tissues during the dosing interval without compromising the overall PK profile and addressing the need for large dose q.d treatment.

Food effect: results part 3:

the PK data for the food effect cohort summarized in tables 2-3 below show a lower rate of absorption as indicated by a 1.25 fold decrease in Cmax and a more complete overall absorption indicated by a 1.4 fold increase in AUC 0-24. Most importantly, the mean Tmax varied from 1 hour (fasted) to >3 hours (fed).

(FIG. 4)

Table 2-3PK parameters summary: food Effect of Compound (I) after a Single dose of 60mg

CV (coefficient of variation) (%) SD/average value 100

Pharmacodynamics

Pharmacodynamics (PD) are characterized by assessing target occupancy and distal pathway inhibition. Measurement of BTK occupancy in human whole blood (given as the ratio of free BTK to total BTK) serves as a direct marker of therapeutic target occupancy.

The relationship between BTK occupancy, dose, systemic compound exposure, and efficacy of complex in vivo pathways and disease readings has been established among various preclinical models of compounds having formula (I). (example 1)

The compounds of formula (I) are irreversible inhibitors of BTK, determining the extent and duration of BTK occupancy. The PD effect of compound (I) was assessed by measuring free BTK (unbound) and total BTK in whole blood by enzyme-linked immunosorbent assay (ELISA) on the Meso Scale Diagnostics (MSD) platform in two separate assays.

The relationship between dose and pharmacodynamics is characterized by measurements of BTK occupancy in human blood (as a ratio of free BTK to total BTK), which is a direct marker of therapeutic target occupancy. BTK occupancy was determined for a single escalating dose ranging from 0.5 to 400mg, multiple escalating doses ranging from 10mg to 400mg, and multiple escalating doses ranging from 100mg to 200 mg.

The compounds of formula (I) exhibit a significant dose-dependent increase in both the extent and duration of peripheral blood BTK occupancy. Peak target occupancy is typically observed at 0.5 hours post-dose, indicating that the drug is rapidly acting without associated hysteresis relative to peak drug exposure. The conclusion drawn from its ability to covalently bind BTK is that target occupancy far exceeds its ability to be disposed of from the systemic circulation, suggesting the existence of an unbalanced PK-PD relationship. Thus, it was concluded that the duration of BTK occupancy was dependent on the de novo synthesis rate of BTK.

Unlike the lower dose group (0.5-1.5mg), a single dose of 15mg and above of compound (I) established near 100% peak target occupancy in almost all subjects and remained above 80% over 24 hours. Although the response varied widely between subjects at 15mg, 30mg and higher doses had sustained (>24h) and near complete (> 90%) occupancy in all subjects, with significantly reduced inter-subject variability. The time to refresh the BTK protein pool to pre-dose levels was about 10 days, corresponding to a median turnover T1/2 of about 48 hours (fig. 5).

After multiple doses of compound (I), 10mg of compound (I) q.d had achieved > 96% BTK occupancy prior to day 12 dosing.

In addition, ex vivo inhibition of basophil activation (monitored by surface expression of CD63 and CD203 c) was used as a remote mechanistic biomarker to test the downstream PD effect of compound (I). To determine the PD effect of compound (I) on basophil activation, whole blood was stimulated ex vivo with anti-IgE. Degranulation was assessed by flow cytometry by the percentage of CD63+ and CD203+ basophils.

Following a single escalating dose of compound (I), the data indicate dose-dependent inhibition of fcer 1-mediated basophil activation. Ex vivo blood basophil activation was almost completely inhibited (> 89%) at the 60mg dose, while reaching close to 100% inhibition 24h after administration at higher doses, as measured by CD 63. In contrast, the maximal inhibition of CD203c (about 50% inhibition) was only achieved on 200mg of compound (I) 24 hours after a single dose of compound (I).

At day 12, 8h after q.d. or b.i.d. administration of MAD with compound of formula (I), the lowest tested dose of compound (I) (10mg qd) had resulted in > 90% inhibition of CD63 upregulation, and the trough level of CD63 was > 90% inhibition at doses of compound (I) of > 50mg q.d. (figure 6). The maximum trough inhibition of CD203c activation at day 12 lasted higher than after a single dose of compound (I) and was only achieved when 100mg and 200mg of compound (I) were administered b.i.d.

In the MAD study part of the first human study, the ability of compound (I) to inhibit defined allergen reactions was evaluated by the Skin Prick Test (SPT). SPT was performed at different time points before dosing (screening, baseline and before dosing on day 1) and after the first dose (day 1) and 11 days after once daily dosing (day 12).

Similar to the inhibition of ex vivo basophil activation, a dose-dependent effect on wheal diameter can be clearly seen in multiple ascending dose cohorts, as indicated by a decrease in mean wheal size after administration compared to baseline (fig. 7). The effect begins to plateau at about 100mg compound (I) q.d.

Reasons/conclusions for dose selection

Healthy volunteers have been exposed to compound (I) in a phase 1 clinical study at doses ranging from 0.5mg to 600mg given in a single dose or up to 18 days daily one or two. Compound (I) is well tolerated and there are no serious or severe adverse events associated with the ingestion of compound (I). In the clinical study, observed Adverse Events (AEs) did not appear to be dose-dependent, were mostly single-events, and were generally of lesser nature. Therefore, clinical safety information supports the dose selected for this phase 2b study.

The dose levels of the invention were derived from the following assays (BTK occupancy, inhibition of basophil activation (monitored by CD63 and CD203c upregulation)) in healthy volunteers; and a surrogate analysis of the effect of skin needle test (SPT) in asymptomatic specific healthy volunteers-inhibition of mast cells and basophils in the skin.

In the above clinical trial, q.d. administration of 10mg of compound (I) resulted in almost complete occupancy of BTK in the blood, a > 90% decrease in CD63 upregulation (8 hours after compound (I) administration in steady state), and minimal inhibition of wheal size in SPTs. Thus, 10mg of compound (I) q.d. corresponds to the onset of biological activity. At 100mg of compound of formula (I), the average decrease in wheal size in the SPT begins to level off. Thus, 100mg of the compound corresponds to the maximum effect of compound (I). A q.d. moderate dose of thirty-five mg of compound (I) is well suited to accurately describe the dose-response curve for q.d. compound (I).

Compounds having formula (I) inhibit BTK by covalent binding. When BTK occupancy in blood is >24 hours (h), rapid turnover of BTK in tissues (e.g., given in rodent spleen for approximately 5 hours) may require b.i.d. administration of compound (I) for maximum efficacy. The doses of compound (I) of 10mg, 25mg and 100mg b.i.d. accurately describe the dose response curves of compound (I) given twice daily, respectively.

Security in humans

To analyze adverse reactions, placebo subjects from all SAD and MAD groups (2 per group) and subjects divided by SAD and MAD section were combined into one placebo group (SAD n-20 and MAD n-16) for comparison with each single compound (I) dose group (n-6 each) and total compound (I) group (SAD n-60 and MAD n-48). There were no significant major differences in demographic data between placebo and active groups for the SAD and MAD populations. Safety assessments conducted in the FIH study in healthy volunteers did not show significant safety concerns at doses up to 600 mg.

Example 3: efficacy and safety data for moderate to severe SjS patients

A phase 2 study was conducted on the compound of formula (I) with the aim of determining safety and efficacy and characterizing the dose response of the compound of formula (I) in subjects with moderate to severe SjS in order to further develop the compound for the treatment of this disease.

Summary of the schemes

Target and endpoint

TABLE 2-4 targets and associated endpoints

Design of research

This is an adaptive phase 2 randomized, double-blind, placebo-controlled, multi-center, integrated dose range study aimed at evaluating the safety and efficacy of multiple doses of a compound of formula (I) in patients with moderate to severe sjogren's syndrome (SjS). In this study, moderate to severe SjS was defined as sjogren's syndrome according to the ACR/EULAR criteria, and ESSDAI was at least 5 (8 out of 12 domains) and ESSPRI was at least 5. If the study subjects received some concomitant treatment for the underlying disease and still met the entry criteria, they would continue to receive such treatment, provided that they remained stable until the end of the study.

The study consisted of two parts. In part 1 of this study, the single dose of highest biological activity tested in two different dosing regimens (100mg of a compound having formula (I) or a pharmaceutically acceptable salt thereof): once daily (qd) or twice daily (bid) and compared to placebo. A total of about 72 subjects were evenly randomly assigned to the 3 treatment groups, with an expected sample size of 24 subjects per group. In part 2, the selected dosing regimen (qd or bid) was extended to lower doses to evaluate the safety and dose response of the dosing regimen between the lowest dose (10mg of the compound of formula (I)) and the highest dose (100mg of the compound of formula (I)) dose found. This resulted in 4 treatment groups; placebo plus three dose levels of compound of formula (I) (100mg bid/qd, 25mg bid or 35mg qd and 10mg bid/qd). A total of about 180 subjects are expected to be evenly randomized to these 4 treatment groups, resulting in a sample size of 45 subjects per group.

Each individual study subject first received a screening period of up to 6 weeks, a treatment duration of 24 weeks, and a follow-up period of 30 days after the last study treatment administration before the study visit was concluded. The total duration of each subject in the study (including the screening period) was up to 35 weeks.

Throughout the duration of the treatment period (24 weeks), the subjects received twice daily administration of the compound of formula (I) or placebo, whichever dosing regimen was selected, in order to maintain blindness throughout the study.

Safety assessments include physical examination, ECG, vital signs, standard clinical laboratory assessments (hematology, blood chemistry, urinalysis), and adverse event and severe adverse event monitoring.

Screening

After signing the informed consent, subjects received ESSDAI and ESSPRI assessments and completed safety and other assessments to assess eligibility. For traffic scheduling reasons, assessments were made at different dates within a 6-week screening period if the investigator deemed appropriate. Subjects who fail the screen may have one opportunity to participate in the re-screen. The subject is also provided with a wearable device and guidance on how to use the wearable device if they have selected to use the wearable device if the subject is confirmed to be eligible to participate in the study.

Base line

Eligible subjects will return for the baseline visit on day 1. Due to traffic scheduling, subjects may be overnight in the study center, but this is not considered hospitalization. Qualification is confirmed prior to randomization and the required baseline assessment should be completed prior to day 1 dosing. Some baseline assessments were performed the evening prior to day 1 if the study center was preferred for scheduling purposes.

Study treatment

The study medication was provided in bottles with appropriate blind-fill labels. These bottles contain capsules containing 10mg or 25mg or 50mg of active substance (compound of formula (I)) or a matching placebo. Each dose (2 capsules) is swallowed with water and should be taken on an empty stomach. Between morning and evening administration, an interval of about 12 hours (10 to 14 hours) should be maintained. Detailed information on storage and management requirements for study treatments, as well as subject numbers, study treatment assignments, and instructions for administration are outlined in SOM.

Treatment group (arm/group)

In part 1, subjects were assigned to one of the following 3 treatment groups at a 1:1:1 ratio on day 1

A compound of formula (I), 100mg twice daily

A compound of formula (I), 100mg qd

Placebo

The dosing schedule and dose range for part 2 is based on IA data. In part 2, subjects were assigned to one of the following 4 treatment groups at a ratio of 1:1:1:1 on day 1

A compound of formula (I), 100mg bid or qd

A compound of formula (I), 35mg qd or 25mg bid

10mg bid or qd of a compound of formula (I)

Placebo

The subject took 2 capsules at each administration. All subjects in both fractions had morning and evening doses (2 capsules each).

Subjects received a morning dose of compound having formula (I) or placebo at the study center on day 1, day 29 (week 4), day 57 (week 8), week 85 (week 12), day 113 (week 16), day 141 (week 20), and day 169 (week 24). Other morning doses and all evening doses are typically taken at home by the subjects taking part in the trial. All subjects received a corresponding supply of compound or placebo capsules of formula (I) every 4 weeks during the scheduled study center visit.

Subjects were randomly assigned to each treatment group per study section. With the exception of Japan, random assignments were stratified by baseline ESSDAI (based on a weighted score of < or > 10). Separate randomly assigned numbers were generated for subjects in japan and other participating countries to ensure that japanese subjects were evenly distributed across all treatment groups of the study. Subjects received a morning dose of compound having formula (I) or placebo at study center on day 1 (week 1), day 29 (week 4), day 57 (week 8), week 85 (week 12), day 113 (week 16), day 141 (week 20), and day 169 (week 24). The subject is then provided with the study medication and the subject may return home to continue their daily dosing regimen (self-administration).

Subjects also received post-dose safety and PK assessments at week 4 and week 24 visits. Subjects returned to the study center approximately every 4 weeks, with additional visits at the end of week 2 (day 15). In the study visit, subjects received ESSDAI and ESSPRI assessments as well as other scale/questionnaires, safety and various PK, PD and biomarker sample collections as indicated in the assessment schedule.

Subjects were asked to complete a weekly diary to record their SjS symptoms and administration of treatment.

The primary endpoint of the study was assessed after completion of 24 weeks of treatment at the end of part 2 (day 169, end of week 24 visit). The interim analysis evaluates efficacy and safety after 12 weeks of treatment as a surrogate for 24 week treatment outcomes in section 1.

Rationale for dose/regimen and duration of treatment

Part 1 dosage/regimen

Based on the predicted BTK occupancy in blood (B-cell block) and tissue and inhibition of CD63 upregulation In Basophils (IB) (results in example 2), the highest dose (100mgbid or qd) of this study plan has shown the greatest effect of the compound of formula (I). Thus, it can be concluded that this dose can provide the greatest clinical effect against SjS in tissues including lymphoid tissues. In phase 1, doses up to 600mg in single and multiple administrations, and 200mg in twice daily administrations have been tested in human volunteers and proved to be safe.

In part 1, the dose that provides the greatest effect (100mg) was tested in the qd and bid regimens and compared to placebo. Due to the covalent nature of the binding of the compound of formula (I) to intracellular BTK, the duration of therapeutic efficacy depends on the turnover rate of the BTK molecule. The simulation model showed that at steady state, the qd regimen containing 100mg of the compound of formula (I) provided an average BTK occupancy of 83% at 24 hours after dosing (and immediately before the next dose), whereas the bid regimen with the same single dose of 100mg had an average BTK occupancy of 96% at 24 hours.

Furthermore, 70% BTK inhibition at steady state for about 90% of each dosing period is considered appropriate for optimal clinical efficacy. Thus, the qd and bid regimens tested in part 1 of this study, both of which are 100mg of compound (I) dosing regimens, may provide efficacy.

Part 2 dosage/regimen

Part 2 will evaluate the complete dosage range of the selected dosing regimen of between 10mg and 100 mg. In the first human study with the compound of formula (I), q.d. administration of 10mg of the compound of formula (I) resulted in almost complete BTK occupancy in the blood and a > 80% reduction in CD63 upregulation, but only minimal inhibition of wheal size in the skin needle test (SPT). It was therefore concluded that: 10mg of compound q.d. having formula (I) will correspond to the onset of the biological activity of the pharmacokinetic activity in the tissue. As an intermediate dose, a daily dose of 35mg qd of a compound of formula (I) or 25mg bid is considered to accurately describe the complete dose response curve for compound qd or bid of formula (I), respectively. A double-blind treatment period lasting up to 24 weeks will provide sustained safety and efficacy data for compounds having formula (I).

Reasons for control drug (placebo) selection

The comparative treatment was a placebo providing objective evidence of potential AE and other safety data as well as clinical efficacy and PD data generated by subjects treated with a compound of formula (I) or a pharmaceutically acceptable salt thereof during the 24 week trial. The use of placebo is reasonable because there is no approved SjS systemic treatment. Current standard of care for SjS patients is limited to symptomatic care for mucosal signs and symptoms (dryness), and steroids and conventional DMARDs are often ineffective. No pharmaceutical intervention was effective against the severe disabling fatigue associated with SjS.

46页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:激酶抑制剂的用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!