US20210106584A1 - Methods for treating immune thrombocytopenia by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile - Google Patents

Methods for treating immune thrombocytopenia by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile Download PDF

Info

Publication number
US20210106584A1
US20210106584A1 US17/069,218 US202017069218A US2021106584A1 US 20210106584 A1 US20210106584 A1 US 20210106584A1 US 202017069218 A US202017069218 A US 202017069218A US 2021106584 A1 US2021106584 A1 US 2021106584A1
Authority
US
United States
Prior art keywords
treatment period
prior
human patient
itp
platelet count
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/069,218
Other languages
English (en)
Inventor
Steven GOURLAY
Ann Neale
Philip NUNN
Claire LANGRISH
Olga Bandman
Dolca Thomas
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Principia Biopharma Inc
Original Assignee
Principia Biopharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Principia Biopharma Inc filed Critical Principia Biopharma Inc
Priority to US17/069,218 priority Critical patent/US20210106584A1/en
Publication of US20210106584A1 publication Critical patent/US20210106584A1/en
Assigned to PRINCIPIA BIOPHARMA INC. reassignment PRINCIPIA BIOPHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GOURLAY, Steven, BANDMAN, OLGA, NUNN, PHILIP, THOMAS, Dolca, LANGRISH, Claire, NEALE, Ann
Priority to US17/946,585 priority patent/US20230124267A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents

Definitions

  • BTK inhibitors and pharmaceutical compositions comprising the same are also disclosed.
  • Immune thrombocytopenia is a rare autoimmune disease that causes high risk for bleeding, excessive bruising, and fatigue, as well as the potential for life threatening intracranial bleeding due to destruction of platelets. ITP is characterized by immune-mediated (e.g., autoantibody-mediated) platelet destruction and impaired platelet production, resulting in thrombocytopenia, a predisposition to bleeding associated with morbidity and mortality, and adverse impact on patient quality of life (QOL).
  • immune-mediated e.g., autoantibody-mediated
  • ITP intravenous immunoglobulin
  • corticosteroids intravenous immunoglobulin
  • TPO-RAs thrombopoietin receptor agonists
  • rituximab thrombopoietin receptor agonists
  • fostamatinib rituximab
  • other immunosuppressive therapies such as, e.g., mycophenolate mofetil (MMF) and cyclosporine.
  • pharmacotherapy e.g., corticosteroids, IVIG, or anti-D immunoglobulin therapy
  • pharmacotherapy is used for symptomatic patients with low platelet counts for reducing platelet destruction. While most patients respond initially to corticosteroids, the rate of continued remission is low.
  • Second line therapies for ITP include rituximab and splenectomy, which are associated with risk of sepsis and immune suppression. Additionally, thrombopoietin (TPO) mimetics (Bussel 2007) are approved for the treatment of patients with chronic ITP who have not had sufficient responses to corticosteroids, IVIG, or splenectomy.
  • TPO thrombopoietin
  • Novel, safe, and effective oral treatments to maintain platelet counts in ITP patients would represent a significant therapeutic advantage over current standard of care.
  • unmet needs in relapsed and refractory ITP include: improving remission rates and durability; avoiding rapid increase of platelet counts/thrombosis risk; steroid-free regimens; and a tolerable and safe therapy that ensures good patient QOL.
  • novel oral therapies for treating ITP including relapsed and refractory ITP, that address some or all of these limitations of existing therapeutic modalities.
  • BTK Bruton's agammaglobulinemia tyrosine kinase
  • BCR B-cell receptor
  • Fc ⁇ R Fc-gamma receptor
  • Fc ⁇ R Fc-epsilon receptor
  • BTK is a non-receptor tyrosine kinase and a member of the TEC family of kinases.
  • BTK is essential to B cell lineage maturation, and inhibition of BTK activity in cells produces phenotypic changes consistent with blockade of the BCR.
  • BTK inhibition results in the down-regulation of various B-cell activities, including cell proliferation, differentiation, maturation, and survival, and the up-regulation of apoptosis.
  • BTK may be best viewed as an immune function “modulator” (Crofford L J et al., 2016; Pal Singh S et al., 2018).
  • Important insights into BTK function come from loss of function analyses in humans and mice.
  • Individuals with loss of function mutations in the BTK gene develop X-linked agammaglobulinemia (XLA), characterized by a complete absence of circulating B cells and plasma cells, and very low levels of immunoglobulins of all classes (Tsukada 1993, Vetrie 1993). This indicates the potential for BTK inhibition to suppress production of autoantibodies thought to be important in the development of autoimmune diseases, such as, e.g., ITP.
  • XLA X-linked agammaglobulinemia
  • BTK is not expressed in T cells, natural killer cells, and plasma cells and has no traceable direct functions in T cells and plasma cells (Sideras and Smith 1995; Mohamed et al., 2009), the enzyme regulates the activation of other hematopoietic cells, such as basophils, mast cells, macrophages, neutrophils, and platelets.
  • BTK plays a role in the activation of neutrophils, which are key players in the inflammatory response that contributes to wound healing but may also cause tissue damage (Volmering S et al., 2016).
  • a selective BTK inhibitor has the potential to target multiple pathways involved in inflammation and autoimmunity, including, but not limited to: blocking BCR; inhibiting plasma cell differentiation and antibody production; blocking IgG-mediated Fc ⁇ R activation, phagocytosis, and inflammatory mediators in monocytes or macrophages; blocking IgE-mediated Fc ⁇ R activation and degranulation in mast cells or basophils; and inhibiting activation, adhesion, recruitment, and oxidative burst in neutrophils. Based on these effects, a selective BTK inhibitor may block the initiation and progression of various inflammatory diseases and mitigate tissue damage resulting from these diseases.
  • BTKi BTK inhibitors
  • PCI-32765 ibrutinib
  • spebrutinib CC-292
  • BTKi BTK inhibitors
  • PCI-32765 ibrutinib
  • spebrutinib spebrutinib
  • Ibrutinib has also demonstrated activity in other hematological malignancies (Wang 2013, Byrd 2013, Imbruvica Package Insert, 2015).
  • CC-292 has been reported to be well tolerated in a healthy volunteer population at doses which provide 100% occupancy of the BTK enzyme (Evans 2013). Furthermore, evobrutinib recently demonstrated efficacy for multiple sclerosis in a Phase 2 trial (Montalban X et al., 2019).
  • BTKi compounds are in clinical development for various immune-mediated disorders, such as pemphigus (NCT02704429), rheumatoid arthritis (NCT03823378, NCT03682705, NCT03233230), and asthma (NCT03944707) (Montalban X et al., 2019; Norman P 2016; Tam C S et al., 2018; Crawford J J et al., 2018; Min T K et al., 2019; Gillooly K M 2017; Nadeem A et al., 2019).
  • BTKi covalent BTKi
  • ibrutinib and acalabrutinib improved on the selectivity issues that plagued many first-generation kinase inhibitors
  • these inhibitors are typically irreversible, causing permanent modification of both on- and off-target kinases and side effects such as thrombocytopenia, anemia, platelet aggregation, and hepatotoxicity (RITUXAN Prescribing Information, 2018; Drug Record Kinase Inhibitors, 2019; Khan Y et al., 2019; Paydas S, 2019; IMBRUVICA, 2013; Rigg R A et al., 2016; Tang C P S et al., 2018).
  • immune-mediated diseases such as, e.g., ITP, based on BTKi with reduced side effects.
  • Compound (I) is a BTK inhibitor of the following structure:
  • PRN1008 is also known as PRN1008 and rilzabrutinib.
  • This compound has been disclosed in several patent publications, such as, e.g., PCT Publication Nos. WO 2014/039899, WO 2015/127310, WO 2016/100914, WO 2016/105531, and WO 2018/005849, the contents of each of which are incorporated by reference herein.
  • PRN1008 is a novel, highly selective, small molecule inhibitor of non-T cell white blood cell signaling via B-cell receptor, FC ⁇ R, and/or Fc ⁇ R signaling of the BTK pathway.
  • PRN1008 functions as a reversible covalent BTK inhibitor and forms both a non-covalent and a covalent bond with its target, allowing for enhanced selectivity and extended inhibition with low systemic exposure.
  • PRN1008 has shown minimal cross-reactivity with other molecules and is low risk for off-target effects (Smith P F et al., 2017).
  • PRN1008's reversible binding minimizes the likelihood of permanently modified peptides (Serafimova I M 2012).
  • PRN1008 shows improved kinase selectivity relative to the covalent BTK inhibitor ibrutinib, with PRN1008 (1 ⁇ M) achieving >90% inhibition for 6 kinases compared to 21 kinases for ibrutinib (1 ⁇ M) in a 251-kinase panel.
  • PRN1008 has shown encouraging results for the treatment of immune-mediated diseases.
  • PRN1008 is the most advanced BTKi in development for an autoimmune disease (Phase 3, NCT03762265) and the first BTKi to be evaluated in the treatment of pemphigus, a blistering disease that, like ITP, is autoantibody-driven.
  • PRN1008 is rapidly absorbed following oral administration, with a fast half-life (3-4 h) and variable pharmacokinetics (PK).
  • PRN1008 has also demonstrated a favorable safety profile. Based on preclinical reproductive toxicity studies, PRN1008 is not expected to harm fetal development or male fertility. In a Phase 1 study in healthy volunteers, the most commonly reported adverse events were gastrointestinal adverse events, including nausea/vomiting and diarrhea. No serious adverse events or deaths were reported, and no participants discontinued treatment due to an adverse event (Smith P F 2017).
  • PRN1008 demonstrates a significant dose dependent reduction of platelet-loss (consumption) in a mouse model of immune thrombocytopenia.
  • PRN1008 also shows rapid and significant anti-inflammatory effects in a rat collagen-induced arthritis model, a rat antibody-mediated arthus model, spontaneous canine pemphigus foliaceus, and human pemphigus vulgaris (PV).
  • ITP immune thrombocytopenia
  • methods for treating immune thrombocytopenia (ITP) in a human patient in need thereof comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • R immune thrombocytopenia
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise treating primary ITP. In some embodiments, the methods comprise treating secondary ITP. In some embodiments, the methods comprise treating chronic ITP. In some embodiments, the methods comprise treating relapsing ITP. In some embodiments, the methods comprise treating refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • ITP immune thrombocytopenia
  • the methods comprise increasing at least 2 consecutive platelet counts relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 2 consecutive platelet counts relative to a pre-administration baseline platelet count without requiring rescue medication.
  • the methods comprise increasing a platelet count by at least 5,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing a platelet count by at least 10,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing a platelet count by at least 15,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing a platelet count by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 30% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 40% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 50% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 60% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 70% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 6 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 7 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period. In some embodiments, the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period.
  • consecutive platelet counts are measured at least 5 days apart.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • Also disclosed herein are methods for achieving a platelet count of at least 50,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 30% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 35% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 40% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 45% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 55% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 60% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 65% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 70% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 6 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 7 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 6 platelet counts measured during the treatment period.
  • the methods comprise increasing at least 30% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 40% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 50% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 60% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 70% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 6 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 7 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period. In some embodiments, the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period.
  • consecutive platelet counts are measured at least 5 days apart.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • Also disclosed herein are methods for achieving at least one platelet count of at least 50,000/ ⁇ L and increasing at least one platelet count by at least 20,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 30% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 35% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 40% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 45% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 55% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 60% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 65% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 70% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 6 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 7 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 6 platelet counts measured during the treatment period.
  • consecutive platelet counts are measured at least 5 days apart.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • Also disclosed herein are methods for achieving a platelet count of at least 30,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • some embodiments of the disclosure include:
  • a method for treating immune thrombocytopenia (ITP) in a human patient in need thereof comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period, wherein the human patient has at least one characteristic chosen from:
  • a method for increasing a platelet count in a human patient with immune thrombocytopenia comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period, wherein the human patient has at least one characteristic chosen from:
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period. 7.
  • a method for achieving a platelet count of at least 50,000/ ⁇ L in a human patient with immune thrombocytopenia comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period, wherein the human patient has at least one characteristic chosen from:
  • achieving a platelet count comprises achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period.
  • a method for achieving at least one platelet count of at least 50,000/ ⁇ L and increasing at least one platelet count by at least 20,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period, wherein the human patient has at
  • achieving a platelet count comprises achieving at least 2 platelet counts of at least 50,000/ ⁇ L.
  • the method according to Embodiment 13 or 14, wherein achieving a platelet count comprises achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L.
  • the method according to any of Embodiments 13-15, wherein achieving a platelet count comprises achieving a platelet count of at least 50,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period.
  • achieving a platelet count comprises achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 8 platelet counts measured during the treatment period. 18.
  • achieving a platelet count comprises achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period. 19.
  • the method according to any of Embodiments 13-18, wherein achieving a platelet count comprises increasing at least 50% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period. 21.
  • a method for achieving a platelet count of at least 30,000/ ⁇ L in a human patient with immune thrombocytopenia comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period, wherein the human patient has at least one characteristic chosen from:
  • achieving a platelet count comprises achieving a platelet count of at least 30,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period
  • achieving a platelet count comprises achieving a platelet count of at least 30,000/ ⁇ L in at least 75% of platelet counts measured during the treatment period
  • the at least one concomitant ITP therapy is chosen from corticosteroids and thrombopoietin receptor agonists. 50. The method according to Embodiment 48 or 49, wherein the at least one concomitant ITP therapy is chosen from corticosteroids, eltrombopag, and romiplostim. 51.
  • the at least one compound comprises at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof. 52.
  • the at least one compound comprises at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof. 53.
  • the at least one compound comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the at least one compound is orally administered to the human patient.
  • some embodiments of the disclosure include:
  • a method for treating immune thrombocytopenia in a human patient comprising: administering to the patient a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008).
  • PRN1008 comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile.
  • PRN1008 is formulated as a pharmaceutical composition.
  • the pharmaceutical composition comprises one or more pharmaceutical acceptable carriers or excipients. 5.
  • Embodiment 4 wherein the pharmaceutical composition comprises microcrystalline cellulose, crospovidone, and sodium stearyl fumarate with a film coating. 6.
  • the patient is administered an initial dose of 400 mg QD of PRN1008, and after 28 days, the dose is escalated to a higher dose of 300 mg BID or 400 mg BID of PRN1008.
  • the method according to Embodiment 6, wherein the treatment period is for a minimum of 8 days.
  • the method according to Embodiment 6, wherein the treatment period ranges from 8 days to 28 days.
  • the patient's platelet count is less than 30,000/ ⁇ L for two or more consecutive platelet counts. 15.
  • the patient has a stable response of a platelet count of greater than or equal to 50,000/ ⁇ L during the treatment period and an increase of platelet count of ⁇ 20,000/ ⁇ L from baseline.
  • 22. The method according to Embodiment 1, wherein before administration, the patient has relapsed or refractory idiopathic thrombocytopenia, which is primary or secondary to other diseases afflicting the patient.
  • 23. The method according to Embodiment 1, wherein in the patient has secondary ITP. 24.
  • Embodiment 21 wherein the patient is administered a dose of 400 mg BID.
  • the patient is also taking one or more concomitant medications.
  • the one or more concomitant medication is chosen from corticosteroids, eltrombopag, and romiplostim. 27.
  • a method for treating immune thrombocytopenia in a human patient comprising: administering to the patient a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]-pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008), wherein before administration, the patient's platelet count is less than 30,000/ ⁇ L for two or more consecutive platelet counts, and wherein after administration, the patient has a stable response of a platelet count of greater than or equal to 50,000/ ⁇ L during the treatment period.
  • QD once daily
  • BID 300 mg twice daily
  • BID 400 mg BID of 2-[(3R)-3-[4-amino-3-(
  • a method for treating immune thrombocytopenia in a human patient comprising: administering to the patient a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]-pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008), wherein before administration, the patient's platelet count is less than 30,000/ ⁇ L for two or more consecutive platelet counts, and after administration over a treatment period, the patient obtains two or more consecutive platelet counts, separated by at least 5 days, of ⁇ 50,000/ ⁇ L and an increase of platelet count of ⁇ 20,000/ ⁇ L
  • FIG. 1A illustrates PRN1008's reversible covalent binding mechanism, which enables optimized clinical activity with minimal drug exposure and benefits associated with reversibility.
  • FIG. 1B depicts PK exposure (ng/mL) and % BTK inhibition over 25 h for PRN1008.
  • FIG. 2 depicts the dose-escalation rules for the dose-finding portion of an adaptive, open-label, dose-finding, phase 1/2 study investigating PRN1008 in ITP patients.
  • FIG. 3A depicts the effects of 1 ⁇ M PRN1008 on platelet function as assessed in vitro in normal healthy volunteer blood platelets using a standard panel of platelet agonists.
  • FIG. 3B depicts the effects of 1 ⁇ M PRN1008 on platelet function as assessed in vitro in ITP patient blood platelets using a standard panel of platelet agonists.
  • FIG. 4 depicts the effects of 1 ⁇ M ibrutinib on platelet function as assessed in vitro in normal healthy volunteer blood platelets using a standard panel of platelet agonists.
  • FIG. 5 depicts the platelet count response over time for a patient with secondary ITP enrolled in a phase 1/2 study investigating PRN1008 in ITP patients.
  • FIG. 8 depicts summary statistics for platelet response in phase 1/2 ITP patients (data cut-off: Nov. 13, 2019).
  • FIG. 10 depicts summary statistics for platelet response in phase 1/2 ITP patients (data cut-off: Apr. 22, 2020).
  • a or “an” entity refers to one or more of that entity; for example, a compound refers to one or more compounds or at least one compound unless stated otherwise. As such, the terms “a” (or “an”), “one or more”, and “at least one” can be used interchangeably herein.
  • the term “about” is used herein to mean approximately, in the region of, roughly, or around. When the term “about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 5%. With regard to specific values, it should be understood that specific values described herein for subject populations (e.g., the subject of the described clinical trial) represent median, mean, or statistical numbers, unless otherwise provided. Accordingly, aspects of the present disclosure requiring a particular value in a subject are supported herein by population data in which the relevant value is assessed to be a meaningful delimitation on the subject population.
  • active pharmaceutical ingredient or “therapeutic agent” (“API”) refers to a biologically active compound.
  • administer refers to providing, giving, dosing, and/or prescribing by either a health practitioner or an authorized agent and/or putting into, taking or consuming by the patient or person himself or herself.
  • administration of an API to a patient refers to any route (e.g., oral delivery) of introducing or delivering the API to the patient. Administration includes self administration and administration by another.
  • BID and “bid” are used interchangeably to refer to twice a day.
  • ITP immune thrombocytopenia
  • short acute
  • chronic long term
  • ITP affects multiple age groups and can be seen in children, teenagers, and adults.
  • the term “in combination with,” when referring to two or more compounds, agents, or additional active pharmaceutical ingredients, means the administration of two or more compounds, agents, or active pharmaceutical ingredients to the patient prior to, concurrent with, or subsequent to each other during a treatment period.
  • the two or more compounds, agents, or active pharmaceutical ingredients may be administered on different schedules during the treatment period, such as, e.g., with one or more compounds, agents, or active pharmaceutical ingredients being administered once a day and one or more other compounds, agents, or active pharmaceutical ingredients being administered twice a day.
  • an amount expressed in terms of “mg of [X]” refers to the total amount in milligrams of [X], i.e., the free base.
  • PRN1008 may be administered as a pharmaceutically acceptable salt of PRN1008, in which case an amount expressed in terms of “mg of PRN1008” refers to the total amount in milligrams of PRN1008, i.e., the free base, plus the equivalent amount of one or more pharmaceutically acceptable salts of PRN1008 based on the weight of free base therein.
  • “400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof” includes 400 mg of PRN1008 and a concentration of one or more pharmaceutically acceptable salts of PRN1008 equivalent to 400 mg of PRN1008.
  • a “pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, and neither biologically nor otherwise undesirable, such as, e.g., a carrier or an excipient that is acceptable for mammalian pharmaceutical use.
  • the term “pharmaceutically acceptable salt” refers to a salt form, e.g., an acid addition salt, of an active pharmaceutical agent that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the API of which the salt is made.
  • Pharmaceutically acceptable salts are well known in the art and include those derived from suitable inorganic and organic acids.
  • Such salts include, but are not limited to, salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, benzenesulfonic acid, 4-toluenesulfonic acid, and the like.
  • S. M. Berge et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19.
  • PRN1008 As used herein, the terms “PRN1008,” “rilzabrutinib,” “(R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile” and “2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]-pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile” are used interchangeably to refer to a compound having the structure:
  • a dose of PRN1008 may contain the corresponding (S) enantiomer as an impurity in less than about 5% by weight, such as, e.g., as an impurity in less than about 1% by weight.
  • a dose of the (E) isomer of PRN1008 may contain the corresponding (Z) isomer as an impurity in less than about 1% by weight;
  • a dose of the (Z) isomer of PRN1008 may contain the corresponding (E) isomer as an impurity in less than about 1% by weight.
  • PRN1008 is denoted as a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile, it means that the amount of (E) or (Z) isomer in the mixture is greater than about 1% by weight. In some embodiments, the molar ratio of (E) to (Z) isomer is 9:1.
  • PRN1008 or a pharmaceutically acceptable salt thereof may also be referred to herein as a “drug,” “active agent,” “a therapeutically active agent,” or “API.”
  • the term “therapeutically effective amount” refers to that an of a compound that produces the desired effect for which it is administered (e.g., improvement in ITP or a symptom of ITP, or lessening the severity of ITP or a symptom of ITP).
  • the exact amount of an effective dose will depend on the purpose of the treatment and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • the term “treat,” “treating,” or “treatment,” when used in connection with a disorder or condition includes any effect, e.g., lessening, reducing, modulating, ameliorating, or eliminating, that results in the improvement of the disorder or condition. Improvements in or lessening the severity of any symptom of the disorder or condition can be readily assessed according to standard methods and techniques known in the art.
  • Some embodiments of the present disclosure relate to methods for treating immune thrombocytopenia (ITP) in a human patient in need thereof comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise treating primary ITP. In some embodiments, the methods comprise treating secondary ITP. In some embodiments, the methods comprise treating chronic ITP. In some embodiments, the methods comprise treating relapsing ITP. In some embodiments, the methods comprise treating refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the human patient has at least 2 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 3 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 4 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 5 characteristics prior to the start of the treatment period chosen from:
  • the human patient has all of the following characteristics prior to the start of the treatment period:
  • the human patient is from 21 to 74 years old prior to the start of the treatment period.
  • the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the human patient had ITP for at least 2 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 months prior to the start of the treatment period.
  • the human patient had ITP for at least 1 year prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 2 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 7 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 8 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 9 years prior to the start of the treatment period.
  • the human patient had ITP for at least 10 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 20 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 30 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 40 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 50 years prior to the start of the treatment period.
  • the human patient has a history of taking from 1 to 41 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking from 1 to 54 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least one prior ITP therapy prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 2 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 3 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 4 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 5 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 6 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 7 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 8 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 9 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 10 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 15 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 20 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 25 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 30 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 35 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 40 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 45 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 50 prior ITP therapies prior to the start of the treatment period.
  • the at least one prior ITP therapy is chosen from corticosteroids, thrombopoietin receptor agonists, intravenous immunoglobulin, anti-D immunoglobulin, and rituximab.
  • the human patient had a splenectomy prior to the start of the treatment period.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • the at least one concomitant ITP therapy is chosen from corticosteroids and thrombopoietin receptor agonists. In some embodiments, the at least one concomitant ITP therapy is chosen from corticosteroids, eltrombopag, and romiplostim.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • Some embodiments of the present disclosure relate to methods for increasing a platelet count in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the methods comprise increasing at least 2 consecutive platelet counts relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 2 consecutive platelet counts relative to a pre-administration baseline platelet count without requiring rescue medication.
  • the methods comprise increasing a platelet count by at least 5,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing a platelet count by at least 10,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing a platelet count by at least 15,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing a platelet count by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 30% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 40% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 50% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 60% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 70% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 6 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 7 of the final 8 platelet counts measured during the treatment period by at least 20,000/O_, relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 6 platelet counts measured during the treatment period by at least 20,000/O_, relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period. In some embodiments, the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period.
  • consecutive platelet counts are measured at least 5 days apart.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the human patient has at least 2 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 3 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 4 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 5 characteristics prior to the start of the treatment period chosen from:
  • the human patient has all of the following characteristics prior to the start of the treatment period:
  • the human patient is from 21 to 74 years old prior to the start of the treatment period.
  • the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the human patient had ITP for at least 2 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 months prior to the start of the treatment period.
  • the human patient had ITP for at least 1 year prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 2 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 7 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 8 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 9 years prior to the start of the treatment period.
  • the human patient had ITP for at least 10 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 20 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 30 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 40 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 50 years prior to the start of the treatment period.
  • the human patient has a history of taking from 1 to 41 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking from 1 to 54 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least one prior ITP therapy prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 2 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 3 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 4 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 5 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 6 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 7 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 8 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 9 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 10 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 15 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 20 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 25 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 30 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 35 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 40 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 45 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 50 prior ITP therapies prior to the start of the treatment period.
  • the at least one prior ITP therapy is chosen from corticosteroids, thrombopoietin receptor agonists, intravenous immunoglobulin, anti-D immunoglobulin, and rituximab.
  • the human patient had a splenectomy prior to the start of the treatment period.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • the at least one concomitant ITP therapy is chosen from corticosteroids and thrombopoietin receptor agonists. In some embodiments, the at least one concomitant ITP therapy is chosen from corticosteroids, eltrombopag, and romiplostim.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • Some embodiments of the present disclosure relate to methods for achieving a platelet count of at least 50,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 30% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 35% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 40% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 45% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 55% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 60% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 65% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 70% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 6 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 7 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 6 platelet counts measured during the treatment period.
  • the methods comprise increasing at least 30% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 40% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 50% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 60% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 70% of platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 6 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 7 of the final 8 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the methods comprise increasing at least 2 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 3 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 4 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count. In some embodiments, the methods comprise increasing at least 5 of the final 6 platelet counts measured during the treatment period by at least 20,000/ ⁇ L relative to a pre-administration baseline platelet count.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period. In some embodiments, the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period.
  • the pre-administration baseline platelet count is an average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period.
  • consecutive platelet counts are measured at least 5 days apart.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the human patient has at least 2 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 3 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 4 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 5 characteristics prior to the start of the treatment period chosen from:
  • the human patient has all of the following characteristics prior to the start of the treatment period:
  • the human patient is from 21 to 74 years old prior to the start of the treatment period.
  • the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the human patient had ITP for at least 2 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 months prior to the start of the treatment period.
  • the human patient had ITP for at least 1 year prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 2 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 7 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 8 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 9 years prior to the start of the treatment period.
  • the human patient had ITP for at least 10 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 20 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 30 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 40 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 50 years prior to the start of the treatment period.
  • the human patient has a history of taking from 1 to 41 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking from 1 to 54 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least one prior ITP therapy prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 2 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 3 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 4 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 5 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 6 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 7 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 8 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 9 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 10 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 15 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 20 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 25 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 30 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 35 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 40 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 45 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 50 prior ITP therapies prior to the start of the treatment period.
  • the at least one prior ITP therapy is chosen from corticosteroids, thrombopoietin receptor agonists, intravenous immunoglobulin, anti-D immunoglobulin, and rituximab.
  • the human patient had a splenectomy prior to the start of the treatment period.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • the at least one concomitant ITP therapy is chosen from corticosteroids and thrombopoietin receptor agonists. In some embodiments, the at least one concomitant ITP therapy is chosen from corticosteroids, eltrombopag, and romiplostim.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • Some embodiments of the present disclosure relate to methods for achieving at least one platelet count of at least 50,000/ ⁇ L and increasing at least one platelet count by at least 20,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L. In some embodiments, the methods comprise achieving at least 2 consecutive platelet counts of at least 50,000/ ⁇ L without requiring rescue medication.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 30% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 35% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 40% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 45% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 55% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 60% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 65% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 70% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 6 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 7 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 2 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 3 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 50,000/ ⁇ L in at least 5 of the final 6 platelet counts measured during the treatment period.
  • consecutive platelet counts are measured at least 5 days apart.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the human patient has at least 2 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 3 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 4 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 5 characteristics prior to the start of the treatment period chosen from:
  • the human patient has all of the following characteristics prior to the start of the treatment period:
  • the human patient is from 21 to 74 years old prior to the start of the treatment period.
  • the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the human patient had ITP for at least 2 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 months prior to the start of the treatment period.
  • the human patient had ITP for at least 1 year prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 2 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 7 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 8 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 9 years prior to the start of the treatment period.
  • the human patient had ITP for at least 10 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 20 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 30 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 40 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 50 years prior to the start of the treatment period.
  • the human patient has a history of taking from 1 to 41 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking from 1 to 54 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least one prior ITP therapy prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 2 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 3 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 4 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 5 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 6 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 7 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 8 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 9 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 10 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 15 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 20 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 25 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 30 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 35 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 40 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 45 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 50 prior ITP therapies prior to the start of the treatment period.
  • the at least one prior ITP therapy is chosen from corticosteroids, thrombopoietin receptor agonists, intravenous immunoglobulin, anti-D immunoglobulin, and rituximab.
  • the human patient had a splenectomy prior to the start of the treatment period.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • the at least one concomitant ITP therapy is chosen from corticosteroids and thrombopoietin receptor agonists. In some embodiments, the at least one concomitant ITP therapy is chosen from corticosteroids, eltrombopag, and romiplostim.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • Some embodiments of the present disclosure relate to methods for achieving a platelet count of at least 30,000/ ⁇ L in a human patient with immune thrombocytopenia (ITP) comprising administering to the human patient a therapeutically effective amount of at least one compound chosen from (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008) and pharmaceutically acceptable salts thereof once a day or twice a day for a treatment period.
  • ITP immune thrombocytopenia
  • the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 30% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 35% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 40% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 45% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 50% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 55% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 60% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 65% of platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 70% of platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 2 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 3 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 4 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 5 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 6 of the final 8 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 7 of the final 8 platelet counts measured during the treatment period.
  • the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 2 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 3 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 4 of the final 6 platelet counts measured during the treatment period. In some embodiments, the methods comprise achieving a platelet count of at least 30,000/ ⁇ L in at least 5 of the final 6 platelet counts measured during the treatment period.
  • the treatment period is 8 days. In some embodiments, the treatment period is 28 days. In some embodiments, the treatment period is 84 days. In some embodiments, the treatment period is 168 days.
  • the treatment period is at least 8 days. In some embodiments, the treatment period is at least 28 days. In some embodiments, the treatment period is at least 84 days. In some embodiments, the treatment period is at least 168 days.
  • the treatment period is from 8 days to 28 days. In some embodiments, the treatment period is from 8 days to 84 days. In some embodiments, the treatment period is from 8 days to 168 days. In some embodiments, the treatment period is from 28 days to 84 days. In some embodiments, the treatment period is from 28 days to 168 days. In some embodiments, the treatment period is from 84 days to 168 days.
  • the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day or twice a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof once a day. In some embodiments, the methods comprise administering to the human patient 400 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the methods comprise administering to the human patient 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof twice a day.
  • the human patient has primary ITP. In some embodiments, the human patient has secondary ITP. In some embodiments, the human patient has chronic ITP. In some embodiments, the human patient has relapsing ITP. In some embodiments, the human patient has refractory ITP.
  • the human patient has at least one characteristic prior to the start of the treatment period chosen from:
  • the human patient has at least 2 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 3 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 4 characteristics prior to the start of the treatment period chosen from:
  • the human patient has at least 5 characteristics prior to the start of the treatment period chosen from:
  • the human patient has all of the following characteristics prior to the start of the treatment period:
  • the human patient is from 21 to 74 years old prior to the start of the treatment period.
  • the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has 2 platelet counts of less than 30,000/ ⁇ L prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart. In some embodiments, the human patient has a platelet count of less than 30,000/ ⁇ L for at least 2 consecutive platelet counts prior to the start of the treatment period, wherein the 2 counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 28,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 15,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L. In some embodiments, the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the average of the human patient's two most recent platelet counts prior to the start of the treatment period, wherein the two counts are measured no sooner than 7 days apart in the 15 days prior to the start of the treatment period, and the human patient's platelet count on the first day of the treatment period is from 3,000/ ⁇ L to 33,000/ ⁇ L.
  • the human patient had ITP for at least 2 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 months prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 months prior to the start of the treatment period.
  • the human patient had ITP for at least 1 year prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 2 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 3 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 4 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 5 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 6 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 7 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 8 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 9 years prior to the start of the treatment period.
  • the human patient had ITP for at least 10 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 20 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 30 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 40 years prior to the start of the treatment period. In some embodiments, the human patient had ITP for at least 50 years prior to the start of the treatment period.
  • the human patient has a history of taking from 1 to 41 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking from 1 to 54 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least one prior ITP therapy prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 2 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 3 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 4 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 5 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 6 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 7 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 8 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 9 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 10 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 15 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 20 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 25 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 30 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 35 prior ITP therapies prior to the start of the treatment period.
  • the human patient has a history of taking at least 40 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 45 prior ITP therapies prior to the start of the treatment period. In some embodiments, the human patient has a history of taking at least 50 prior ITP therapies prior to the start of the treatment period.
  • the at least one prior ITP therapy is chosen from corticosteroids, thrombopoietin receptor agonists, intravenous immunoglobulin, anti-D immunoglobulin, and rituximab.
  • the human patient had a splenectomy prior to the start of the treatment period.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered as a monotherapy.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof is administered in combination with at least one concomitant ITP therapy.
  • the at least one concomitant ITP therapy is chosen from corticosteroids and thrombopoietin receptor agonists. In some embodiments, the at least one concomitant ITP therapy is chosen from corticosteroids, eltrombopag, and romiplostim.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof comprises a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (E) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of at least one compound chosen from the (Z) isomer of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile and pharmaceutically acceptable salts thereof.
  • the at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof consists of a mixture of (E) and (Z) isomers of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile or a pharmaceutically acceptable salt of the foregoing.
  • the present disclosure is directed to a method for treating immune thrombocytopenia in a human patient comprising: administering to the patient a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]-pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008).
  • QD once daily
  • BID 300 mg twice daily
  • BID 400 mg BID of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]-pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-
  • PRN1008 comprises a mixture of (E) and (Z) isomers of 2-[(3R)-3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]-pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile.
  • PRN1008 is formulated as a pharmaceutical composition and the pharmaceutical composition comprises one or more pharmaceutical acceptable carriers or excipients.
  • the pharmaceutical composition comprises microcrystalline cellulose, crospovidone, and sodium stearyl fumarate with a film coating.
  • the method further comprises repeating the administration of PRN1008 to the patient over a treatment period. Additionally, in some embodiments, the treatment period ranges from 28 days to 168 days, for a minimum of 8 days or from 8 days to 28 days.
  • the present disclosure provides that before administration, the patient's platelet count is less than 30,000/ ⁇ L for two or more consecutive platelet counts. Further, in some embodiments, after administration over a treatment period, the patient obtains two or more consecutive platelet counts, separated by at least 5 days, of ⁇ 50,000/ ⁇ L the treatment period ranges from 28 days to 168 days. Additionally, in some embodiments, after administration over a treatment period, the patient has a stable response of a platelet count of greater than or equal to 50,000/ ⁇ L during the treatment period and an increase of platelet count of ⁇ 20,000/ ⁇ L from baseline.
  • the present disclosure provides that before administration, the patient has relapsed or refractory idiopathic thrombocytopenia, which is primary or secondary to other diseases afflicting the patient.
  • the patient has secondary ITP.
  • the patient is administered a dose of 400 mg QD.
  • the present disclosure is directed to the occurrence where the patient is also taking one or more concomitant medications.
  • the one or more concomitant medication is chosen from corticosteroids, eltrombopag, and romiplostim.
  • the present disclosure is directed to a method for treating immune thrombocytopenia in a human patient comprising: administering to the patient a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008), wherein before administration, the patient's platelet count is less than 30,000/ ⁇ L for two or more consecutive platelet counts, and wherein after administration, the patient has a stable response of a platelet count of greater than or equal to 50,000/ ⁇ L during the treatment period.
  • a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of (R)-2-
  • the present disclosure is directed to a method for treating immune thrombocytopenia in a human patient comprising: administering to the patient a dose chosen from 400 mg once daily (QD), 300 mg twice daily (BID), and 400 mg BID of (R)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile (PRN1008), wherein before administration, the patient's platelet count is less than 30,000/ ⁇ L for two or more consecutive platelet counts, and after administration over a treatment period, the patient obtains two or more consecutive platelet counts, separated by at least 5 days, of ⁇ 50,000/ ⁇ L and an increase of platelet count of ⁇ 20,000/ ⁇ L from baseline.
  • PRN1008 is administered as part of a pharmaceutical composition comprising: at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; and at least one pharmaceutically acceptable excipient.
  • the pharmaceutical composition is in the form of at least one tablet.
  • PRN1008 is orally administered as part of a pharmaceutical composition comprising: at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; and at least one pharmaceutically acceptable excipient.
  • the pharmaceutical composition is in the form of at least one tablet.
  • the pharmaceutical composition is in the form of at least one tablet comprising 100 mg or 300 mg of PRN1008.
  • the pharmaceutical composition is in the form of at least one tablet comprising 100 mg of PRN1008.
  • the pharmaceutical composition is in the form of at least one tablet comprising 300 mg of PRN1008.
  • PRN1008 is administered in the form of a film-coated tablet.
  • PRN1008 is administered in the form of at least one tablet comprising: at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; and at least one pharmaceutically acceptable excipient.
  • PRN1008 is administered in the form of at least one tablet comprising: at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; at least one filler; at least one disintegrant; at least one lubricant; and at least one film coating.
  • the at least one filler is microcrystalline cellulose.
  • the at least one disintegrant is crospovidone.
  • the at least one lubricant is sodium stearyl fumarate.
  • PRN1008 is administered in the form of at least one tablet comprising: 100 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; and at least one pharmaceutically acceptable excipient.
  • PRN1008 is administered in the form of at least one tablet comprising: 100 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; at least one filler; at least one disintegrant; at least one lubricant; and at least one film coating.
  • the at least one filler is microcrystalline cellulose.
  • the at least one disintegrant is crospovidone.
  • the at least one lubricant is sodium stearyl fumarate.
  • PRN1008 is administered in the form of at least one tablet comprising: 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; and at least one pharmaceutically acceptable excipient.
  • PRN1008 is administered in the form of at least one tablet comprising: 300 mg of at least one compound chosen from PRN1008 and pharmaceutically acceptable salts thereof; at least one filler; at least one disintegrant; at least one lubricant; and at least one film coating.
  • the at least one filler is microcrystalline cellulose.
  • the at least one disintegrant is crospovidone.
  • the at least one lubricant is sodium stearyl fumarate.
  • PRN1008 is administered with a glass of water.
  • PRN1008 is administered with food.
  • PRN1008 is administered without food.
  • any pharmaceutically acceptable excipient may be determined by the chosen route of administration and standard pharmaceutical practice. Except insofar as any conventional pharmaceutically acceptable excipient is incompatible with PRN1008, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically composition, its use is contemplated to be within the scope of this disclosure.
  • materials which may serve as pharmaceutically acceptable excipients include: (1) sugars, such as, e.g., lactose, glucose, and sucrose; (2) starches, such as, e.g., corn starch and potato starch; (3) cellulose and its derivatives, such as, e.g., sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as, e.g., cocoa butter and suppository waxes; (9) oils, such as, e.g., peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as, e.g., propylene glycol; (11) polyols, such as, e.g., glycerin, sorbitol, mannito
  • NCT03395210 An ongoing phase 1/2 clinical trial (NCT03395210) investigating the safety, pharmacokinetics, and clinical activity of PRN1008, an oral BTK inhibitor, in patients with relapsed/refractory immune thrombocytopenia (ITP) began enrolling patients on Mar. 22, 2018. As of Oct. 6, 2020, the estimated primary completion date for the study is September 2022, with an estimated study completion date of September 2023. To date, PRN1008 has been well-tolerated in ITP patients with no reported treatment-related bleeding or thrombotic events. Moreover, positive preliminary results have been observed in a highly treatment-resistant and refractory patient population.
  • the key inclusion criteria for the phase 1/2 study are: adults aged 18-80 years old with relapsed/refractory ITP; ITP primary or secondary to other diseases (e.g., systemic lupus erythematosus, chronic lymphocytic leukemia); no other available/approved treatment options; ⁇ 2 platelet counts ⁇ 30,000/ ⁇ L at study entry; and adequate hematologic, hepatic, and renal function.
  • Enrolled patients have low platelet counts, having relapsed on or been refractory to prior therapies with no available and approved therapeutic options and may continue corticosteroids and/or thrombopoietin mimetics during the study.
  • corticosteroids for example, stable concomitant corticosteroid (CS) or thrombopoietin receptor agonist (TPO-RA) treatment is permitted during the study.
  • TPO-RA thrombopoietin receptor agonist
  • sample size employed in the study is based on clinical considerations with the intention of gaining a sufficiently high confidence level in the study results using normal approximation methods.
  • the completed PRN1008 intrapatient dose escalation portion of the study used a 3+3 design. Specifically, if response was observed in 1 of 3 patients, then 3 more patients were added to the dose level. However, if no response was observed in 3 patients for 28 days, the dose was dropped, and patients were escalated to higher doses and all subsequently enrolled patients started treatment at the higher dose.
  • patients were orally dosed with PRN1008 for an active treatment period of 24 weeks at doses of 200 and 400 mg qd and 300 and 400 mg bid. If patients exhibited a response at an initial dose, the patient continued at that dose ( FIG. 2 ). If the patient did not respond to the initial dose, the dose was escalated to a higher dose.
  • CBCs complete blood counts
  • PK samples were collected intensively on the first day of each new, higher dose level and at random times following dosing at each on-treatment follow up visit to assess dose response.
  • Other standard clinical and laboratory assessments of patients with ITP were also employed in the dose escalation study, including a bleeding assessment (ITP-BAT; Rodeghiero 2013) that has been used in prior ITP trials.
  • the safety objective of the phase 1/2 study is to characterize the safety and tolerability of up to four dose levels of PRN1008 in patients with ITP.
  • safety is assessed by the incidence, severity, and relationship of TEAEs, including clinically significant changes in physical examination, laboratory tests, and vital signs.
  • Adverse events are categorized as treatment emergent after the first dose of PRN1008 has been received.
  • TEAEs in the post-treatment follow-up period are also assessed and examined for possible relationship to the prior PRN1008 treatment.
  • TEAEs include all AEs that start on or after the first dose of study medication, or AEs that are present prior to the first dose of study medication, but their severity or relationship increases after the first dose of study medication up to and including the final study medication dosing date. Within each preferred term, patients are counted only once if they had more than one event reported during the dosing period.
  • the pharmacokinetic objective of the study is to characterize the pharmacokinetics of PRN1008 in patients with ITP.
  • Plasma PK parameters (C max , T max , AUC, t 1/2 , V/F, CL/F) of PRN1008 in ITP patients are evaluated in each patient based on frequent sampling on Day 1 of a new, higher dosing level and reported by dose and, if relevant, overall.
  • Non-compartmental analysis is employed to derive PK parameters for each individual. Results are reported by descriptive statistics, and exploratory analyses may pool these data with the data from other studies of PRN1008.
  • Efficacy objectives of the study include: (1) to explore the clinical activity of up to four dose levels of PRN1008 in relapsed/refractory patients with ITP (200 and 400 mg qd; 300 and 400 mg bid); and (2) to identify a potential dose regimen to use in future studies of PRN1008 in patients with ITP.
  • the dose-finding/dose escalation portion of the study has been completed, and all enrolled patients are currently being treated with 400 mg bid.
  • the primary efficacy outcome measure is consecutive increased platelet counts, i.e., two or more consecutive platelet counts ⁇ 50,000/ ⁇ L without requiring rescue medication.
  • Part A examines the proportion of patients able to achieve two or more consecutive platelet counts, separated by at least 5 days, of ⁇ 50,000/ ⁇ L and an increase of platelet count of ⁇ 20,000/ ⁇ L from baseline, without use of rescue medication in the 4 weeks prior to the latest elevated platelet count.
  • the analysis of the primary efficacy endpoint is a weighted logistic regression, with the binary indicator of platelet response as the dependent variable, and dose level in total mg per day as a covariate, weighted by the number of days the patient was on the dose.
  • Additional efficacy endpoints include: any 2 platelet counts ⁇ 50,000/ ⁇ L; platelet responses over time, by duration of treatment, and clinical benefit ( ⁇ 30,000/ ⁇ L); stable response (platelet counts ⁇ 50,000/ ⁇ L at 50% of visits for 4 of the last 8 weeks of active treatment).
  • secondary efficacy endpoints of the study include: (1) proportion of patients able to achieve two or more platelet counts of ⁇ 50,000/ ⁇ L and increase of platelet count of ⁇ 20,000/ ⁇ L from baseline at any time (on treatment or during follow up) without use of rescue medication in the 4 weeks prior to the latest elevated platelet count; (2) proportion of patients able to achieve two or more platelet counts, separated by at least 5 days, representing an increase of platelet count of ⁇ 20,000/ ⁇ L from baseline, by dose level, without use of rescue medication in the 4 weeks prior to the latest elevated platelet count; (3) proportion of patients able to achieve two or more platelet counts, separated by at least 5 days, of ⁇ 100,000/ ⁇ L, by dose level, without use of rescue medication in the 4 weeks prior to the latest elevated platelet count; (4) change from baseline to the average of the last two platelet counts at each dosing level; (5) time to first platelet response (as defined in the primary endpoint); (6) proportion of patients receiving rescue medication at each dosing level and overall; (7) proportion of patients
  • Exploratory objectives of the study include: the effect of PRN1008 on platelet autoantibody levels; the effect of PRN1008 on markers of hemolysis; the effect of PRN1008 on thrombopoietin (TPO) levels; the effect of PRN1008 on quality of life (QoL) using the Euro-QoL 5-Dimension Visual Analog Scale (EQ-5D VAS); and plasma metabolite analysis/identification for PRN1008.
  • An adaptive, open-label, dose-finding study using rule-based intrapatient dose escalation is an appropriate phase 1/2 design to evaluate the safety and effect of PRN1008 in patients with ITP, a relatively rare autoimmune disease.
  • the study design is similar to that used to study the initial efficacy and safety of the orally available tyrosine kinase inhibitor fostamatinib in ITP (Podolanczuk et al., 2009).
  • the 200 mg qd human starting dose was expected to result in minimal improvement in platelet counts.
  • a 10 mg/kg/d dose was associated with mean 51% BTK occupancy 1 h post-dose, along with a slight increase in platelet count over vehicle control (mean 83.1% platelet decrease from baseline for vehicle with anti-platelet challenge, compared to a 71.9% platelet decrease for PRN1008 10 mg/kg/d).
  • the top dose of 40 mg/kg/d in that model resulted in a maximum BTK occupancy of 91%, matching the expected peak occupancies with the maximum 400 mg QD and 400 mg BID doses.
  • the 400 mg BID dose was expected to produce higher trough, or pre-dose BTK occupancies of approximately 70% vs. 50% for QD dosing, while having similar peak values. Dosing above 400 mg BID is thought unlikely to result in greater target inhibition.
  • the blistering disease pemphigus is an autoantibody-driven disease like ITP.
  • Study PRN1008-005 a rapid onset of clinical effect was seen in most pemphigus patients within 4 weeks, with continued improvement to very low levels of skin inflammation by the end of 12 weeks of therapy. Therefore, 28-day cycles to assess the initial platelet response and trigger intrapatient dose-escalation were deemed appropriate for studying ITP. Additionally, a 24-week overall treatment period was preferred for the dose-finding study to enable all patients to potentially dose-escalate to the higher doses and be fully evaluated at those doses.
  • the DLT evaluation period for any patient is defined as the duration of PRN1008 dosing.
  • Dose-limiting toxicity in the phase 1/2 study, including the completed dose escalation study is established as follows. Hematologic DLT is determined based on: ANC ⁇ 500/ ⁇ L for ⁇ 5 days; Grade 3 or higher decreased hemoglobin in the absence of a pre-existing Grade 2 decreased hemoglobin; febrile neutropenia, with absolute neutrophil count (ANC) ⁇ 1000/mm 3 and single temperature>38.3 degrees C. (101 degrees F.) or a sustained temperature of ⁇ 38° C. (100.4° F.) for more than one hour (CTCAE, version 4.0); and ⁇ Grade 3 or higher bleeding event requiring platelet transfusion.
  • Non-hematologic DLT is considered any >Grade 3 non-hematologic toxicity per the NCI Common Terminology Criteria for Adverse Events (CTCAE), version 4.0, with the following exceptions: laboratory TEAEs that are asymptomatic and return to baseline or to Grade 1 within 7 days; fatigue; nausea, vomiting or diarrhea that return to baseline or Grade 1 within 7 days; and systemic reactions (such as, e.g., fever, headache) that return to baseline or Grade 1 within 7 days. Any toxicity that, at the discretion of the Investigator, is thought to warrant withholding the study drug for more than 7 days is also considered a DLT.
  • CCAE Common Terminology Criteria for Adverse Events
  • n indicates the expected enrollment in each cohort unless a sustained platelet response was seen or a DLT, in which case 3 extra patients would be added to that group. A starting dose level could be dropped for futility after 3 or 6 patients were evaluated, or retained if efficacy is observed. Notably, individual patients in each cohort do not dose-escalate when there is a platelet response at a lower dose level or toxicity. If several dose levels are therapeutic, some or all patients would not reach the higher dose levels.
  • level level** dose dose dose Cohort 4 weeks 4 weeks 4 weeks 4 weeks level** level** 1 200 mg QD 400 mg 300 mg 400 mg 400 mg 400 mg 400 mg (3-6) QD BID BID BID BID 2 400 mg QD 300 mg 400 mg 400 mg 400 mg 400 mg ( ⁇ 6) BID BID BID BID BID 3 300 mg BID 400 mg 400 mg 400 mg 400 mg 400 mg ( ⁇ 6) BID BID BID BID BID 4 400 mg BID 400 mg 400 mg 400 mg 400 mg 400 mg 400 mg 400 mg 400 mg 400 mg ( ⁇ 6) BID BID BID BID BID BID BID BID BID BID BID BID BID BID BID
  • Each patient enrolled in the dose-escalation portion of the study was allowed to up-titrate their dose after 28 days of PRN1008 therapy at each dose level if they did not experience a platelet response (as defined in the primary endpoint) or a DLT at the last dose level. If they experienced a platelet response in the first cycle at any one dose level but did not have a platelet response in the second cycle at that dose, they could dose escalate at the end of the second cycle. Patients experiencing a platelet response (as defined for the primary endpoint) did not have their dose escalated at the next cycle.
  • Patients could receive PRN1008 treatment for up to 24 weeks, starting on Day 1 and ending on Study Day 169, followed by 4 weeks of post-treatment safety follow-up. However, patients who dose escalated to 400 mg BID could continue in the active treatment period until 24 weeks of treatment at the 400 mg BID dose were completed.
  • LTE Long Term Extension
  • LTE Long Term Extension
  • All patients can receive, but are not required to be taking, concomitant corticosteroids.
  • the dose should be fixed ( ⁇ 10%) for at least 2 weeks before Day 1 and remain unchanged throughout the study unless rescue criteria are triggered. If the patient requires rescue treatment or concomitant ITP drug increases of more than 10% of the Day 1 daily dose, the patient will be discontinued from the study and receive rescue treatment per standard of care. These drugs may not have their dose increased as part of “rescue” medication.
  • All patients may receive, but are not required to be taking, eltrombopag or romiplostim.
  • the dose should be fixed for at least 2 weeks before Day 1 and remain constant (10% variation from Day 1 daily dose is allowed) throughout the study unless there are safety concerns related to those drugs. These drugs may not have their dose increased as part of “rescue” medication.
  • PRN1008 is a substrate of P-gp and CYP3A, and an inhibitor of CYP3A.
  • PRN1008 has been shown to act as a moderate CYP3A inhibitor, increasing midazolam exposure by approximately 3-fold.
  • Moderate to strong inducers and inhibitors of cytochrome P450 3A (CYP3A) should be avoided during the study, as they may reduce or increase the exposure of PRN1008 when administered concomitantly.
  • clinically relevant CYP3A substrate drugs with a narrow therapeutic window are not permitted as PRN1008 is a weak to moderate CYP3A inhibitor.
  • Other “sensitive substrates” if they are medically necessary for the patient should be monitored as concurrent use with PRN1008 will increase the blood levels of sensitive substrate drugs.
  • Proton pump inhibitors are not permitted during the phase 1/2 study as they may reduce the bioavailability of PRN1008 tablets.
  • Co-administration of esomeprazole reduced exposure of the tablet formulation of PRN1008 by 48% in a prior study, demonstrating an impact of gastric pH on absorption. Patients could switch to H2 receptor blocking drugs as a substitute.
  • PRN1008 should be administered 2 hours or more prior to permitted acid-reducing drugs.
  • the following inclusion criteria are used to inform the enrollment of patients in the phase 1/2 study, including, e.g., the dose escalation study.
  • exclusion criteria are used to inform the enrollment of patients in the phase 1/2 study, including, e.g., the dose escalation study.
  • participant must fulfill all entry criteria to be enrolled into the study. Participants who fail to meet the entry criteria may be rescreened once at the discretion of the Investigator after informing the study Medical Monitor.
  • subjects After providing informed written consent, subjects typically complete the following clinical assessments: physical examination; medical history; concomitant medications; weight; height; vital signs; ITP-BAT bleeding scale; QOL assessment EQ-5D VAS; online cognitive testing; and safety assessments.
  • Laboratory assessments may be performed at both central and local laboratories, if required.
  • Safety assessments include the following: the frequency, severity and relationship of AEs; clinical laboratory test changes; physical examination, ECGs, vital signs, and cognitive function.
  • PRN1008 is administered in the form a film-coated tablet.
  • PRN1008 tablets are packaged in white high-density polyethylene (HDPE) bottles with child-resistant induction-sealed caps; these bottles are intended to be stored at 2-8° C. and can be transported without ice at room temperature. Additionally, the bottles can be kept at room temperature conditions for up to 2 weeks.
  • HDPE white high-density polyethylene
  • Each PRN1008 film-coated tablet contains either 100 mg or 300 mg of PRN1008 drug substance.
  • the tablet contains Microcrystalline Cellulose (filler), Crospovidone (disintegrant), Sodium Stearyl Fumarate (lubricant), and a non-functional film coating.
  • a 100 mg tablet is a round shape and orange in color.
  • a 300 mg tablet is an oval shape and white in color.
  • PRN1008 tablets should be taken with a glass ( ⁇ 8 oz) of water but may be taken with or without food, i.e., a period of fasting is not required.
  • the Screening Population for this study includes all participants who provide informed consent and have screening assessments evaluated for study participation are included in the Screening Population.
  • the Safety Population includes all participants who have received at least one dose of PRN1008 will be included in the safety analysis.
  • the Safety Population will be used for all safety analyses.
  • 3 evaluable patients defined as compliance of ⁇ 75% of doses for that dose level, are required.
  • patients will be replaced if necessary, to fulfill this requirement.
  • the Intent-to-Treat Exposed (ITT-E) Population includes all participants who have received at least one dose of PRN1008.
  • the Pharmacokinetic Analysis Population includes all participants who have received at least one dose of PRN1008 and have at least one plasma concentration value will be included in the PK analysis.
  • the Pharmacokinetic Analysis Population will be used for all PK analyses. Participants prematurely discontinued from the study, for reasons other than TEAEs, may be replaced at the discretion of the Sponsor to ensure adequate numbers of evaluable participants.
  • a patient who withdraws from the study before the planned end of study visit is considered to have withdrawn from the study early. Participants in this study have the right to withdraw at any time for any reason. Additionally, investigators may withdraw participants from the study in the event of intercurrent illness, AEs, treatment failure after a prescribed procedure, lack of compliance with the study and/or study procedures, or any other reasons where they feel it is in the best interest of the participant to be terminated from the study.
  • IDSM Independent Data Safety Monitor
  • SMC Safety Monitoring Committee
  • CA Competent Authority
  • the IDSM made “sentinel cohort” safety evaluations.
  • the “sentinel patients” for each dose level had their data reviewed by the IDSM, in order to choose the starting dose for additional, new patients. After review, the IDSM could determine that a starting dose for new patients should be dropped for futility (lack of platelet response), increased to the next planned dosing level, kept the same, or reduced.
  • the AE Collection Period begins at the time of the first screening/eligibility assessment and ends at the end of the study for each patient.
  • An AE is any untoward medical occurrence in a participant or clinical investigation participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with the intervention.
  • An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom, or disease temporally associated with the use of an investigational product, whether or not considered related to the product.
  • Investigators are instructed to record all AEs encountered during the clinical study in detail from the date of participant consent throughout the study follow-up period. Pre-existing conditions that worsen during a study are reported as AEs.
  • Investigators are instructed to use their knowledge of the study participant, the circumstances surrounding the event, and an evaluation of any potential alternative causes to determine whether an AE may be considered as related to the study drug, indicating “yes” or “no” accordingly.
  • Investigators are asked to consider following information in assessing relatedness: (1) temporal relationship of event onset to the initiation of study drug; (2) course of the event, considering especially the effects of dose reduction, discontinuation of study drug, or reintroduction of study drug (if applicable); (3) known association of the event with the study drug or with similar treatments; (4) known association of the event with the disease under study; (5) presence of risk factors in the study participant or use of concomitant medications known to increase the occurrence of the events; and (6) presence of non-treatment-related factors that are known to be associated with the occurrence of the event.
  • laboratory and ECG abnormalities accompanied by clinical symptoms, leading to a change in study drug (e.g., dose modification, interruption or permanent discontinuation), or requiring a change in concomitant therapy (e.g., addition of, interruption of, discontinuation of, or any other change in a concomitant medication, therapy or treatment) should be recorded as an AE, with any laboratory or ECG result abnormality fulfilling the criteria for a serious adverse event (SAE) reported as such, in addition to being recorded as an AE.
  • SAE serious adverse event
  • Grade 1 Mild; asymptomatic or mild symptoms; clinical or diagnostic observations only; intervention not indicated.
  • Grade 2 Moderate; minimal, local or noninvasive intervention indicated; limiting age-appropriate instrumental activities of daily living.
  • Grade 3 Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self-care activities of daily living.
  • a serious adverse event is any experience (clinical AE or abnormal laboratory test) that suggests a significant hazard, contraindication, side effect, or precaution.
  • An SAE must fulfill at least one of the following criteria at any dose level: is fatal (results in the outcome death); is life-threatening; requires in-patient hospitalization or prolongation of existing hospitalization; results in persistent or significant disability/incapacity; is a congenital anomaly/birth defect; or is medically significant or requires intervention to prevent one or other of the outcomes listed above.
  • Investigators are instructed to report life-threatening events or any event with an outcome of death should be reported as an SAE.
  • a complete physical examination consists of checking the normality or abnormality of the following body systems: general appearance, skin, eyes, ears, nose, throat, heart, chest/breast, abdomen, neurological system, lymph nodes, spine and extremities (skeletal) and the conduct of and online cognitive testing of learning and memory.
  • An abbreviated physical examination consists of checking the normality or abnormality of the following body systems: general appearance, skin, abdomen, and cardiorespiratory examination. Height is recorded at screening only. Blood pressure (BP), pulse rate, body temperature and respiratory rate are recorded at specific time points.
  • BP Blood pressure
  • ECG assessments are also obtained at specific time points to confirm eligibility and to ensure real time safety evaluation of the participants in the study.
  • participants For ECG evaluations, participants should be in a resting position for at least 10 minutes prior to any measurement. Body position should also be consistently maintained for each ECG evaluation. In particular, changes in heart rate should be avoided. There should be no environmental distractions (TV, radio, conversation) during the pre-ECG rest and the ECG recording time.
  • Heart rate (HR), QRS duration and respiratory rate (RR), and QT intervals are recorded. Changes of the T-wave and U-wave morphology and overall ECG interpretation are documented. All ECG recordings are performed using a standard high-quality, high-fidelity digital electrocardiograph machine equipped with computer-based interval measurements. For triplicate ECG assessments, at least three interpretable ECG recordings (without artifacts) are collected per time point within a ⁇ 10 minute period per time point.
  • Laboratory assessments are performed at a central laboratory, with the provision for occasional local laboratory testing, if required. Laboratory safety tests are collected at specific time points. Additional blood or urine samples may be taken at an investigator's discretion if the results of any test fall outside the reference ranges, or clinical symptoms necessitate additional testing to monitor participant safety. Where the clinical significance of abnormal lab results is considered uncertain, screening lab tests may be repeated before Day 1 to confirm eligibility. In the event of unexplained abnormal clinically significant laboratory test values, the tests should be repeated immediately and followed up until they have returned to the normal range, are considered to be clinically stable, and/or an adequate explanation of the abnormality is found.
  • FIG. 5 shows platelet count response for an enrolled patient with secondary ITP, demonstrating responsiveness to treatment at 400 mg qd.
  • PRN1008 was well-tolerated across all doses (200 and 400 mg qd, 300 and 400 mg bid) in a highly treatment-resistant and refractory patient population with immune thrombocytopenia (ITP). All TEAEs were mild to moderate with no thrombotic events.
  • PRN1008 Phase 1/2 trial of PRN1008 had enrolled 26 adult patients who have had two platelet counts ⁇ 30,000/ ⁇ L within 15 days prior to treatment.
  • Oral PRN1008 starting doses were 200 mg qd, 400 mg qd, 300 mg bid, and 400 mg bid, with intra-patient dose escalation allowed every four weeks, with the trial having a median treatment duration of 12.7 weeks (range: 0.14 to 39.71) at reporting.
  • the 31 enrolled patients in the phase 1/2 study were characterized by the demographic information provided in Tables 4 and 5.
  • the median age of enrolled patients was 50, with 29 of 31 classified with primary ITP (94%) and 2 of 31 classified with secondary ITP (6%).
  • Demographics N 31 Median age, y (range) 50 (21-74) Female, n (%) 18 (58) ITP classification, n (%) Primary ITP 29 (94) Secondary ITP 2 (6) Median duration of ITP, y (range) 7.8 (0.5-42.4) Median baseline platelet count, ⁇ 10 9 /L (range) 13 (3-28)
  • Prior ITP Therapy N 31 Median number of prior ITP therapies (range) 6 (1-41) Splenectomy, n (%) 8 (26) Prior ITP therapies, n (%) Corticosteroids 26 (84) TPO-RA 17 (55) IVIG/Anti-D 11 (35) Rituximab 10 (32)
  • This patient population was highly treatment-resistant and refractory, with patients characterized by a median of 6 prior therapies, including 55% receiving a prior TPO-RA. Rapid onset (platelet counts >30 k by the first week of treatment) was observed following the start of PRN1008 treatment, and responses were durable in the majority of patients. Platelet response was further improved with longer treatment and at higher doses of PRN1008.
  • FIG. 8 Subset analyses of primary platelet responses are depicted in FIG. 8 (data cut-off: Nov. 13, 2019). As of Nov. 13, 2019, 10 of 25 (40%) of heavily pretreated ( ⁇ 4 prior therapies) patients were responding to PRN1008 treatment. Similar responses were achieved in patients receiving PRN1008 monotherapy (4 of 10 patients) and concomitant therapy (8 of 21 patients).
  • PRN1008 was well-tolerated in ITP patients across all doses as of Nov. 13, 2019 (Table 7). The median treatment duration at the Nov. 13, 2019, cut-off was 12.0 weeks (range: 0.1-41.9). Related TEAEs were reported in 11 patients (35%), with all reported TEAEs being grade 1 or 2 (mild to moderate). No treatment-related bleeding or thrombotic events were reported, nor were significant changes in the ITP-BAT bleeding scale from baseline to last visit. No dose-limiting toxicities were observed by data cut-off. The observed safety profile was consistent with the safety observed in pemphigus studies (Murrell D. et al., AAD 2018).
  • platelet responses had a fast onset and were maintained in the majority of patients who started on the PRN1008 400 mg bid dose ( FIG. 9 ).
  • day 8 the first platelet count taken after the start of treatment
  • platelets ⁇ 30 ⁇ 10 9 /L a clinically significant platelet count
  • the primary endpoint was defined as 2 consecutive platelet counts 50,000/ ⁇ L ⁇ without requiring rescue medication.
  • week 4 57% of responders achieved the primary endpoint.
  • 50% of patients achieved the primary endpoint response when initiated on PRN1008 400 mg bid and treated for 12 weeks or more.
  • the response was durable, with responders maintaining platelet counts: 71% of time (weeks) at ⁇ 50 ⁇ 10 9 /L; and 88% of time (week) at ⁇ 20 ⁇ 10 9 /L above baseline.
  • FIG. 10 Subset analyses of platelet responses are depicted in FIG. 10 (data cut-off: Apr. 22, 2020). Overall, 43% of patients met the primary endpoint, which increased with ⁇ 12 weeks of PRN1008. As of Apr. 22, 2020, 15 of 38 (40%) of heavily pretreated ( ⁇ 4 prior therapies) patients were responding to PRN1008 treatment. Similar responses were achieved in patients receiving PRN1008 monotherapy (7 of 16 patients) and inadequate responders on concomitant therapy (13 of 31 patients).
  • PRN1008 was well-tolerated in ITP patients across all doses as of Apr. 22, 2020 (Table 9).
  • the median treatment duration at the Apr. 22, 2020, data cut-off was 17.7 weeks for all patients (range: 0.6-41.9) and 18 weeks for patients initiated on 400 mg bid (range: 1.4-24.6).
  • Related TEAEs were reported in 21 patients (45%), with all reported TEAEs being transient and grade 1 or 2 (mild to moderate). No related serious adverse events were reported. Additionally, no treatment-related bleeding or thrombotic events were reported, nor were significant changes in the ITP-BAT bleeding scale from baseline to last visit. The observed safety profile was consistent with the safety observed in pemphigus studies (Murrell D. et al., AAD 2018).
  • Claims or descriptions that include “or” or “and/or” between at least one members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context.
  • the disclosure includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process.
  • the disclosure includes embodiments in which more than one, or all the group members are present in, employed in, or otherwise relevant to a given product or process.

Landscapes

  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Diabetes (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US17/069,218 2019-10-14 2020-10-13 Methods for treating immune thrombocytopenia by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile Abandoned US20210106584A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/069,218 US20210106584A1 (en) 2019-10-14 2020-10-13 Methods for treating immune thrombocytopenia by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile
US17/946,585 US20230124267A1 (en) 2019-10-14 2022-09-16 Methods for Treating Immune Thrombocytopenia by Administering (R)-2-[3-[4-Amino-3-(2-Fluoro-4-Phenoxy- Phenyl) Pyrazolo[3,4-D]Pyrimidin-1-YL]Piperidine-1-Carbonyl]-4-Methyl-4-[4-(Oxetan-3-YL)Piperazin-1-YL]Pent-2-Enenitrile

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201962914688P 2019-10-14 2019-10-14
US201962951302P 2019-12-20 2019-12-20
US17/069,218 US20210106584A1 (en) 2019-10-14 2020-10-13 Methods for treating immune thrombocytopenia by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/946,585 Continuation US20230124267A1 (en) 2019-10-14 2022-09-16 Methods for Treating Immune Thrombocytopenia by Administering (R)-2-[3-[4-Amino-3-(2-Fluoro-4-Phenoxy- Phenyl) Pyrazolo[3,4-D]Pyrimidin-1-YL]Piperidine-1-Carbonyl]-4-Methyl-4-[4-(Oxetan-3-YL)Piperazin-1-YL]Pent-2-Enenitrile

Publications (1)

Publication Number Publication Date
US20210106584A1 true US20210106584A1 (en) 2021-04-15

Family

ID=73040359

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/069,218 Abandoned US20210106584A1 (en) 2019-10-14 2020-10-13 Methods for treating immune thrombocytopenia by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile
US17/946,585 Pending US20230124267A1 (en) 2019-10-14 2022-09-16 Methods for Treating Immune Thrombocytopenia by Administering (R)-2-[3-[4-Amino-3-(2-Fluoro-4-Phenoxy- Phenyl) Pyrazolo[3,4-D]Pyrimidin-1-YL]Piperidine-1-Carbonyl]-4-Methyl-4-[4-(Oxetan-3-YL)Piperazin-1-YL]Pent-2-Enenitrile

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/946,585 Pending US20230124267A1 (en) 2019-10-14 2022-09-16 Methods for Treating Immune Thrombocytopenia by Administering (R)-2-[3-[4-Amino-3-(2-Fluoro-4-Phenoxy- Phenyl) Pyrazolo[3,4-D]Pyrimidin-1-YL]Piperidine-1-Carbonyl]-4-Methyl-4-[4-(Oxetan-3-YL)Piperazin-1-YL]Pent-2-Enenitrile

Country Status (12)

Country Link
US (2) US20210106584A1 (fr)
EP (1) EP4045051A1 (fr)
JP (1) JP2022550919A (fr)
KR (1) KR20220080179A (fr)
CN (1) CN115209899A (fr)
AU (1) AU2020365951A1 (fr)
CA (1) CA3154257A1 (fr)
CO (1) CO2022006087A2 (fr)
IL (1) IL291994A (fr)
MX (1) MX2022004427A (fr)
TW (1) TW202128175A (fr)
WO (1) WO2021076514A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022221527A1 (fr) * 2021-04-16 2022-10-20 Principia Biopharma, Inc. Procédés de traitement de la thrombocytopénie immunitaire induite par des médicaments et des vaccins par administration de composés spécifiques

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024097667A1 (fr) * 2022-10-31 2024-05-10 Principia Biopharma Inc. Méthodes de traitement de thrombocytopénie immunitaire par administration de (r)-2-[3- [4-amino-3-(2-fluoro-4-phénoxy-phényl)pyrazolo[3,4-d]pyrimidin-l-yl]pipéridine-l-carbonyl]-4-méthyl-4-[4-(oxétan-3-yl)pipérazin-l-yl]pent-2-ènenitrile

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2892900T (pt) 2012-09-10 2017-11-06 Principia Biopharma Inc Compostos de pirazolopirimidina como inibidores de cinase
CN112353806A (zh) 2014-02-21 2021-02-12 普林斯匹亚生物制药公司 Btk抑制剂的盐和固体形式
CN107257686B (zh) 2014-12-18 2021-01-29 普林斯匹亚生物制药公司 天疱疮的治疗
CN107427468B (zh) 2014-12-24 2022-02-25 普林斯匹亚生物制药公司 Btk抑制剂的部位特异性给药
SG11201811255WA (en) 2016-06-29 2019-01-30 Principia Biopharma Inc Modified release formulations of 2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022221527A1 (fr) * 2021-04-16 2022-10-20 Principia Biopharma, Inc. Procédés de traitement de la thrombocytopénie immunitaire induite par des médicaments et des vaccins par administration de composés spécifiques

Also Published As

Publication number Publication date
IL291994A (en) 2022-06-01
CO2022006087A2 (es) 2022-07-29
AU2020365951A1 (en) 2022-06-02
MX2022004427A (es) 2022-07-12
EP4045051A1 (fr) 2022-08-24
KR20220080179A (ko) 2022-06-14
US20230124267A1 (en) 2023-04-20
CA3154257A1 (fr) 2021-04-22
JP2022550919A (ja) 2022-12-05
TW202128175A (zh) 2021-08-01
WO2021076514A1 (fr) 2021-04-22
CN115209899A (zh) 2022-10-18

Similar Documents

Publication Publication Date Title
US20230124267A1 (en) Methods for Treating Immune Thrombocytopenia by Administering (R)-2-[3-[4-Amino-3-(2-Fluoro-4-Phenoxy- Phenyl) Pyrazolo[3,4-D]Pyrimidin-1-YL]Piperidine-1-Carbonyl]-4-Methyl-4-[4-(Oxetan-3-YL)Piperazin-1-YL]Pent-2-Enenitrile
US20190002575A1 (en) Method for the treatment of multiple sclerosis
UA124269C2 (uk) Застосування антагоністу il-13 для лікування атопічного дерматиту
KR20240004451A (ko) Ilt7 결합 단백질을 이용한 자가면역 장애의 치료 방법
CA3229887A1 (fr) Lou064 pour le traitement de la sclerose en plaques
JP7286665B2 (ja) IgE介在アレルギー性疾患の治療
WO2024097667A1 (fr) Méthodes de traitement de thrombocytopénie immunitaire par administration de (r)-2-[3- [4-amino-3-(2-fluoro-4-phénoxy-phényl)pyrazolo[3,4-d]pyrimidin-l-yl]pipéridine-l-carbonyl]-4-méthyl-4-[4-(oxétan-3-yl)pipérazin-l-yl]pent-2-ènenitrile
US20210106583A1 (en) Methods of treating pemphigus by administering (r)-2-[3-[4-amino-3-(2-fluoro-4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-1-yl]piperidine-1-carbonyl]-4-methyl-4-[4-(oxetan-3-yl)piperazin-1-yl]pent-2-enenitrile
RU2800765C2 (ru) ЛЕЧЕНИЕ АЛЛЕРГИЧЕСКИХ ЗАБОЛЕВАНИЙ, ОПОСРЕДОВАННЫХ IgE
US20210380706A1 (en) Methods for treating severe asthma in patients with nasal polyposis
Chronic lr'Jgileau

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

AS Assignment

Owner name: PRINCIPIA BIOPHARMA INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GOURLAY, STEVEN;NEALE, ANN;NUNN, PHILIP;AND OTHERS;SIGNING DATES FROM 20201006 TO 20201014;REEL/FRAME:056802/0501

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION